1
|
Huang J, Chen YL. Zebrafish as a preclinical model for diabetes mellitus and its complications: From monogenic to gestational diabetes and beyond. World J Diabetes 2025; 16:100574. [DOI: 10.4239/wjd.v16.i5.100574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/25/2025] Open
Abstract
With diabetes currently affecting 537 million people globally, innovative research approaches are urgently required. Zebrafish (Danio rerio) has emerged as a pivotal model organism in diabetes research, particularly valuable for developmental biology studies and preclinical therapeutic validation. Its rapid life cycle, optical transparency, and genetic tractability collectively enable efficient longitudinal observation of pathological progression and pharmacological responses. Utilizing zebrafish models, researchers have elucidated fundamental mechanisms governing islet development, β-cell dysfunction, and metabolic dysregulation. These experimental systems have significantly advanced our understanding of various diabetes subtypes, including type 1, type 2, gestational, and monogenic forms, while also facilitating mechanistic studies of diabetic complications such as retinopathy and nephropathy. Recent model refinements, particularly in simulating monogenic disorders and pregnancy-associated metabolic changes, promise to deepen our comprehension of disease pathophysiology and therapeutic interventions. Nevertheless, a persistent limitation lies in their incomplete recapitulation of human-specific physiological complexity and multi-organ metabolic interactions, factors that may influence translational applicability. Despite these constraints, zebrafish-based research continues to provide an indispensable platform for diabetes investigation, holding significant promise for alleviating the escalating global burden of this metabolic disorder.
Collapse
Affiliation(s)
- Jie Huang
- School of Medicine, Hangzhou City University, Hangzhou 310000, Zhejiang Province, China
| | - Yin-Ling Chen
- School of Medicine, Hangzhou City University, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
2
|
Faraj N, Hoogaars WMH, Duinkerken BHP, Wolters AHG, Kats K, Dekkers MC, Zaldumbide A, Giepmans BNG. Pancreatic exocrine damage induces beta cell stress in zebrafish larvae. Diabetologia 2025:10.1007/s00125-025-06432-4. [PMID: 40295334 DOI: 10.1007/s00125-025-06432-4] [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: 10/18/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025]
Abstract
AIMS/HYPOTHESIS Excessive endoplasmic reticulum (ER) stress in beta cells can impair proliferation and contribute to autoimmune responses such as the destruction of beta cells in type 1 diabetes. Exocrine-beta cell interactions affect beta cell growth and function. Notably, exocrine abnormalities are frequently observed alongside overloaded beta cells in different types of diabetes, suggesting that exocrine stress may induce beta cell ER stress and loss. While a cause-consequence relationship between exocrine stress and beta cell function cannot be addressed in humans, it can be studied in a zebrafish model. Larvae develop a pancreas with a human-like morphology by 120 h post-fertilisation, providing a valuable dynamic model for studying pancreatic interactions. Our aim was to target exocrine cells specifically and address beta cell status using transgenic zebrafish models and reporters. METHODS To explore the impact of exocrine damage on beta cell fitness, we generated a novel zebrafish model allowing exocrine pancreas ablation, using a nifurpirinol-nitroreductase system. We subsequently assessed the in vivo effects on beta cells by live-monitoring dynamic cellular events, such as ER stress, apoptosis and changes in beta cell number and volume. RESULTS Exocrine damage in zebrafish decreased pancreas volume by approximately 50% and changed its morphology. The resulting exocrine damage induced ER stress in 60-90% of beta cells and resulted in a ~50% reduction in their number. CONCLUSIONS/INTERPRETATION The zebrafish model provides a robust platform for investigating the interplay between exocrine cells and beta cells, thereby enhancing further insights into the mechanisms driving pancreatic diseases such as type 1 diabetes.
Collapse
Affiliation(s)
- Noura Faraj
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Willem M H Hoogaars
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - B H Peter Duinkerken
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anouk H G Wolters
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kim Kats
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Mette C Dekkers
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ben N G Giepmans
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
3
|
Sharma H, Robea MA, McGrory NH, Bazan DC, Burton EA, Burgess HA. Functional interrogation of neuronal connections by chemoptogenetic presynaptic ablation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647277. [PMID: 40236196 PMCID: PMC11996543 DOI: 10.1101/2025.04.04.647277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Most neurons are embedded in multiple circuits, with signaling to distinct postsynaptic partners playing functionally different roles. The function of specific connections can be interrogated using synaptically localized optogenetic effectors, however these tools are often experimentally difficult to validate or produce paradoxical outcomes. We have developed a system for photoablation of synaptic connections originating from genetically defined neurons, based on presynaptic localization of the fluorogen activating protein dL5** that acts as a photosensitizer when bound to a cell-permeable dye. Using the well mapped zebrafish escape circuit as a readout, we first show that cytoplasmically expressed dL5** enables efficient spatially targeted neuronal ablation using near infra-red light. We then demonstrate that spatially patterned illumination of presynaptically localized dL5** can effectively disconnect neurons from selected downstream partners, producing precise behavioral deficits. This technique should be applicable to almost any genetically tractable neuronal circuit, enabling precise manipulation of functional connectivity within the nervous system.
Collapse
|
4
|
Akhtar MN, Hnatiuk A, Delgadillo-Silva L, Geravandi S, Sameith K, Reinhardt S, Bernhardt K, Singh SP, Maedler K, Brusch L, Ninov N. Developmental beta-cell death orchestrates the islet's inflammatory milieu by regulating immune system crosstalk. EMBO J 2025; 44:1131-1153. [PMID: 39762647 PMCID: PMC11833124 DOI: 10.1038/s44318-024-00332-w] [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: 03/07/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 02/19/2025] Open
Abstract
While pancreatic beta-cell proliferation has been extensively studied, the role of cell death during islet development remains incompletely understood. Using a genetic model of caspase inhibition in beta cells coupled with mathematical modeling, we here discover an onset of beta-cell death in juvenile zebrafish, which regulates beta-cell mass. Histologically, this beta-cell death is underestimated due to phagocytosis by resident macrophages. To investigate beta-cell apoptosis at the molecular level, we implement a conditional model of beta-cell death linked to Ca2+ overload. Transcriptomic analysis reveals that metabolically-stressed beta cells follow paths to either de-differentiation or apoptosis. Beta cells destined to die activate inflammatory and immuno-regulatory pathways, suggesting that cell death regulates the crosstalk with immune cells. Consistently, inhibiting beta-cell death during development reduces pro-inflammatory resident macrophages and expands T-regulatory cells, the deficiency of which causes premature activation of NF-kB signaling in beta cells. Thus, developmental cell death not only shapes beta-cell mass but it also influences the islet's inflammatory milieu by shifting the immune-cell population towards pro-inflammatory.
Collapse
Affiliation(s)
- Mohammad Nadeem Akhtar
- Centre for Regenerative Therapies TU Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus of TU Dresden, German Center for Diabetes Research (DZD e.V.), Dresden, 01307, Germany
- Technische Universität Dresden, CRTD, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstraße 105, 01307, Dresden, Germany
| | - Alisa Hnatiuk
- Centre for Regenerative Therapies TU Dresden, Dresden, 01307, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus of TU Dresden, German Center for Diabetes Research (DZD e.V.), Dresden, 01307, Germany
- Technische Universität Dresden, CRTD, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstraße 105, 01307, Dresden, Germany
| | | | - Shirin Geravandi
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359, Bremen, Germany
| | - Katrin Sameith
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307, Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307, Dresden, Germany
| | - Katja Bernhardt
- Technische Universität Dresden, CRTD, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstraße 105, 01307, Dresden, Germany
| | - Sumeet Pal Singh
- IRIBHM, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
| | - Kathrin Maedler
- Centre for Biomolecular Interactions Bremen, University of Bremen, 28359, Bremen, Germany
| | - Lutz Brusch
- Centre for Interdisciplinary Digital Sciences (CIDS), Information Services and High Performance Computing (ZIH), Technische Universität Dresden, 01187, Dresden, Germany
| | - Nikolay Ninov
- Centre for Regenerative Therapies TU Dresden, Dresden, 01307, Germany.
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital Carl Gustav Carus of TU Dresden, German Center for Diabetes Research (DZD e.V.), Dresden, 01307, Germany.
| |
Collapse
|
5
|
Karampelias C, Liu KC, Tengholm A, Andersson O. Mechanistic insights and approaches for beta cell regeneration. Nat Chem Biol 2025:10.1038/s41589-024-01822-y. [PMID: 39881214 DOI: 10.1038/s41589-024-01822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Diabetes is characterized by variable loss of insulin-producing beta cells, and new regenerative approaches to increasing the functional beta cell mass of patients hold promise for reversing disease progression. In this Review, we summarize recent chemical biology breakthroughs advancing our knowledge of beta cell regeneration. We present current chemical-based tools, sensors and mechanistic insights into pathways that can be targeted to enhance beta cell regeneration in model organisms. We group the pathways according to the cellular processes they affect, that is, proliferation, conversion of other mature cell types to beta cells and beta cell differentiation from progenitor-like populations. We also suggest assays for assessing the functionality of the regenerated beta cells. Although regeneration processes differ between animal models, such as zebrafish, mice and pigs, regenerative mechanisms identified in any one animal model may be translatable to humans. Overall, chemical biology-based approaches in beta cell regeneration give hope that specific molecular pathways can be targeted to enhance beta cell regeneration.
Collapse
Affiliation(s)
- Christos Karampelias
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Ka-Cheuk Liu
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Olov Andersson
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden.
| |
Collapse
|
6
|
Coomer CE, Naumova D, Talay M, Zolyomi B, Snell NJ, Sorkaç A, Chanchu JM, Cheng J, Roman I, Li J, Robson D, McLean DL, Barnea G, Halpern ME. Transsynaptic labeling and transcriptional control of zebrafish neural circuits. Nat Neurosci 2025; 28:189-200. [PMID: 39702668 DOI: 10.1038/s41593-024-01815-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 09/30/2024] [Indexed: 12/21/2024]
Abstract
Deciphering the connectome, the ensemble of synaptic connections that underlie brain function, is a central goal of neuroscience research. Here we report the in vivo mapping of connections between presynaptic and postsynaptic partners in zebrafish, by adapting the trans-Tango genetic approach that was first developed for anterograde transsynaptic tracing in Drosophila. Neural connections were visualized between synaptic partners in larval retina, brain and spinal cord and followed over development. The specificity of labeling was corroborated by functional experiments in which optogenetic activation of presynaptic spinal cord interneurons elicited responses in known motor neuronal postsynaptic targets, as measured by trans-Tango-dependent expression of a genetically encoded calcium indicator or by electrophysiology. Transsynaptic signaling through trans-Tango reveals synaptic connections in the zebrafish nervous system, providing a valuable in vivo tool to monitor and interrogate neural circuits over time.
Collapse
Affiliation(s)
- Cagney E Coomer
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Pharmacology, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Daria Naumova
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Mustafa Talay
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
- Howard Hughes Medical Institute, Department of Molecular and Cellular Biology, Harvard University, Boston, MA, USA
| | - Bence Zolyomi
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Nathaniel J Snell
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Altar Sorkaç
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Jean Michel Chanchu
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, USA
| | - Ji Cheng
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Ivana Roman
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Jennifer Li
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Drew Robson
- Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - David L McLean
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, UK
| | - Gilad Barnea
- Department of Neuroscience, Division of Biology and Medicine, Brown University, Providence, RI, USA
- Robert J. and Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI, USA
| | - Marnie E Halpern
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
7
|
Ceisel A, Emmerich K, McNamara G, Graziano G, Banerjee S, Reibman B, Saxena MT, Mumm JS. Automated In Vivo Phenotypic Screening Platform for Identifying Factors that Affect Cell Regeneration Kinetics. Methods Mol Biol 2025; 2848:217-247. [PMID: 39240526 DOI: 10.1007/978-1-0716-4087-6_14] [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: 09/07/2024]
Abstract
Various strategies for replacing retinal neurons lost in degenerative diseases are under investigation, including stimulating the endogenous regenerative capacity of Müller Glia (MG) as injury-inducible retinal stem cells. Inherently regenerative species, such as zebrafish, have provided key insights into mechanisms regulating MG dedifferentiation to a stem-like state and the proliferation of MG and MG-derived progenitor cells (MGPCs). Interestingly, promoting MG/MGPC proliferation is not sufficient for regeneration, yet mechanistic studies are often focused on this measure. To fully account for the regenerative process, and facilitate screens for factors regulating cell regeneration, an assay for quantifying cell replacement is required. Accordingly, we adapted an automated reporter-assisted phenotypic screening platform to quantify the pace of cellular regeneration kinetics following selective cell ablation in larval zebrafish. Here, we detail a method for using this approach to identify chemicals and genes that control the rate of retinal cell regeneration following selective retinal cell ablation.
Collapse
Affiliation(s)
- Anneliese Ceisel
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, McKusick-Nathans Institute, Human Genetics Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - George McNamara
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gianna Graziano
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shreya Banerjee
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barak Reibman
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Genetic Medicine, McKusick-Nathans Institute, Human Genetics Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Ophthalmology, Center for Nanomedicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
8
|
Almeida LM, Lima LP, Oliveira NAS, Silva RFO, Sousa B, Bessa J, Pinho BR, Oliveira JMA. Zebrafish as a model to study PERK function in developmental diseases: implications for Wolcott-Rallison syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589737. [PMID: 38659860 PMCID: PMC11042256 DOI: 10.1101/2024.04.16.589737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Developmental diseases are challenging to investigate due to their clinical heterogeneity and relatively low prevalence. The Wolcott-Rallison Syndrome (WRS) is a rare developmental disease characterized by skeletal dysplasia and permanent neonatal diabetes due to loss-of-function mutations in the endoplasmic reticulum stress kinase PERK (EIF2AK3). The lack of efficient and less invasive therapies for WRS highlights the need for new animal models that replicate the complex pathological phenotypes, while preserving scalability for drug screening. Zebrafish exhibits high fecundity and rapid development that facilitate efficient and scalable in vivo drug testing. Here, we aimed to assess the potential of zebrafish to study PERK function and its pharmacological modulation, and as model organism of developmental diseases such as the WRS. Using bioinformatic analyses, we showed high similarity between human and zebrafish PERK. We used the pharmacological PERK inhibitor GSK2606414, which was bioactive in zebrafish, to modulate PERK function. Using transgenic zebrafish expressing fluorescent pancreatic markers and a fluorescent glucose probe, we observed that PERK inhibition decreased β cell mass and disrupted glucose homeostasis. By combining behavioural and functional assays, we show that PERK-inhibited zebrafish present marked skeletal defects and defective growth, as well as neuromuscular and cardiac deficiencies, which are clinically relevant in WRS patients, while sparing parameters like otolith area and eye/body ratio which are not associated with WRS. These results show that zebrafish holds potential to study PERK function and its pharmacological modulation in developmental disorders like WRS, assisting research on their pathophysiology and experimental treatments.
Collapse
|
9
|
Karampelias C, Băloiu B, Rathkolb B, da Silva-Buttkus P, Bachar-Wikström E, Marschall S, Fuchs H, Gailus-Durner V, Chu L, Hrabě de Angelis M, Andersson O. Examining the liver-pancreas crosstalk reveals a role for the molybdenum cofactor in β-cell regeneration. Life Sci Alliance 2024; 7:e202402771. [PMID: 39159974 PMCID: PMC11333758 DOI: 10.26508/lsa.202402771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 08/21/2024] Open
Abstract
Regeneration of insulin-producing β-cells is an alternative avenue to manage diabetes, and it is crucial to unravel this process in vivo during physiological responses to the lack of β-cells. Here, we aimed to characterize how hepatocytes can contribute to β-cell regeneration, either directly or indirectly via secreted proteins or metabolites, in a zebrafish model of β-cell loss. Using lineage tracing, we show that hepatocytes do not directly convert into β-cells even under extreme β-cell ablation conditions. A transcriptomic analysis of isolated hepatocytes after β-cell ablation displayed altered lipid- and glucose-related processes. Based on the transcriptomics, we performed a genetic screen that uncovers a potential role of the molybdenum cofactor (Moco) biosynthetic pathway in β-cell regeneration and glucose metabolism in zebrafish. Consistently, molybdenum cofactor synthesis 2 (Mocs2) haploinsufficiency in mice indicated dysregulated glucose metabolism and liver function. Together, our study sheds light on the liver-pancreas crosstalk and suggests that the molybdenum cofactor biosynthesis pathway should be further studied in relation to glucose metabolism and diabetes.
Collapse
Affiliation(s)
- Christos Karampelias
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Bianca Băloiu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Birgit Rathkolb
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Munich, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Patricia da Silva-Buttkus
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Etty Bachar-Wikström
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Susan Marschall
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lianhe Chu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
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
|
11
|
Arefanian H, Al Madhoun A, Al-Rashed F, Alzaid F, Bahman F, Nizam R, Alhusayan M, John S, Jacob S, Williams MR, Abukhalaf N, Shenouda S, Joseph S, AlSaeed H, Kochumon S, Mohammad A, Koti L, Sindhu S, Abu-Farha M, Abubaker J, Thanaraj TA, Ahmad R, Al-Mulla F. Unraveling Verapamil's Multidimensional Role in Diabetes Therapy: From β-Cell Regeneration to Cholecystokinin Induction in Zebrafish and MIN6 Cell-Line Models. Cells 2024; 13:949. [PMID: 38891081 PMCID: PMC11171639 DOI: 10.3390/cells13110949] [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: 03/27/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
This study unveils verapamil's compelling cytoprotective and proliferative effects on pancreatic β-cells amidst diabetic stressors, spotlighting its unforeseen role in augmenting cholecystokinin (CCK) expression. Through rigorous investigations employing MIN6 β-cells and zebrafish models under type 1 and type 2 diabetic conditions, we demonstrate verapamil's capacity to significantly boost β-cell proliferation, enhance glucose-stimulated insulin secretion, and fortify cellular resilience. A pivotal revelation of our research is verapamil's induction of CCK, a peptide hormone known for its role in nutrient digestion and insulin secretion, which signifies a novel pathway through which verapamil exerts its therapeutic effects. Furthermore, our mechanistic insights reveal that verapamil orchestrates a broad spectrum of gene and protein expressions pivotal for β-cell survival and adaptation to immune-metabolic challenges. In vivo validation in a zebrafish larvae model confirms verapamil's efficacy in fostering β-cell recovery post-metronidazole infliction. Collectively, our findings advocate for verapamil's reevaluation as a multifaceted agent in diabetes therapy, highlighting its novel function in CCK upregulation alongside enhancing β-cell proliferation, glucose sensing, and oxidative respiration. This research enriches the therapeutic landscape, proposing verapamil not only as a cytoprotector but also as a promoter of β-cell regeneration, thereby offering fresh avenues for diabetes management strategies aimed at preserving and augmenting β-cell functionality.
Collapse
Affiliation(s)
- Hossein Arefanian
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (R.N.); (S.J.); (S.J.); (L.K.); (T.A.T.)
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Fatema Al-Rashed
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
| | - Fawaz Alzaid
- Department of Bioenergetics & Neurometabolism, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.); (M.R.W.)
- Institut Necker Enfants Malades (INEM), French Institute of Health and Medical Research (INSERM), Immunity & Metabolism of Diabetes (IMMEDIAB), Université de Paris Cité, 75014 Paris, France
| | - Fatemah Bahman
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
| | - Rasheeba Nizam
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (R.N.); (S.J.); (S.J.); (L.K.); (T.A.T.)
| | - Mohammed Alhusayan
- Department of Bioenergetics & Neurometabolism, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.); (M.R.W.)
| | - Sumi John
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (R.N.); (S.J.); (S.J.); (L.K.); (T.A.T.)
| | - Sindhu Jacob
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (R.N.); (S.J.); (S.J.); (L.K.); (T.A.T.)
| | - Michayla R. Williams
- Department of Bioenergetics & Neurometabolism, Dasman Diabetes Institute, Dasman 15462, Kuwait; (F.A.); (M.A.); (M.R.W.)
| | - Nermeen Abukhalaf
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Steve Shenouda
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
| | - Shibu Joseph
- Special Services Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Halemah AlSaeed
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
| | - Shihab Kochumon
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
| | - Anwar Mohammad
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.M.); (M.A.-F.); (J.A.)
| | - Lubaina Koti
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (R.N.); (S.J.); (S.J.); (L.K.); (T.A.T.)
| | - Sardar Sindhu
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
- Animal and Imaging Core Facility, Dasman Diabetes Institute, Dasman 15462, Kuwait;
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.M.); (M.A.-F.); (J.A.)
- Department of Translational Research, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.M.); (M.A.-F.); (J.A.)
| | - Thangavel Alphonse Thanaraj
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (R.N.); (S.J.); (S.J.); (L.K.); (T.A.T.)
| | - Rasheed Ahmad
- Department of Immunology & Microbiology, Dasman Diabetes Institute, Dasman 15462, Kuwait; (H.A.); (F.A.-R.); (F.B.); (S.S.); (H.A.); (S.K.); (S.S.); (R.A.)
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (A.A.M.); (R.N.); (S.J.); (S.J.); (L.K.); (T.A.T.)
| |
Collapse
|
12
|
Chen J, Wang H, Wu S, Zhang A, Qiu Z, Huang P, Qu JY, Xu J. col1a2+ fibroblasts/muscle progenitors finetune xanthophore countershading by differentially expressing csf1a/1b in embryonic zebrafish. SCIENCE ADVANCES 2024; 10:eadj9637. [PMID: 38578990 PMCID: PMC10997200 DOI: 10.1126/sciadv.adj9637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/29/2024] [Indexed: 04/07/2024]
Abstract
Animals evolve diverse pigment patterns to adapt to the natural environment. Countershading, characterized by a dark-colored dorsum and a light-colored ventrum, is one of the most prevalent pigment patterns observed in vertebrates. In this study, we reveal a mechanism regulating xanthophore countershading in zebrafish embryos. We found that csf1a and csf1b mutants altered xanthophore countershading differently: csf1a mutants lack ventral xanthophores, while csf1b mutants have reduced dorsal xanthophores. Further study revealed that csf1a is expressed throughout the trunk, whereas csf1b is expressed dorsally. Ectopic expression of csf1a or csf1b in neurons attracted xanthophores into the spinal cord. Blocking csf1 signaling by csf1ra mutants disrupts spinal cord distribution and normal xanthophores countershading. Single-cell RNA sequencing identified two col1a2+ populations: csf1ahighcsf1bhigh muscle progenitors and csf1ahighcsf1blow fibroblast progenitors. Ablation of col1a2+ fibroblast and muscle progenitors abolished xanthophore patterns. Our study suggests that fibroblast and muscle progenitors differentially express csf1a and csf1b to modulate xanthophore patterning, providing insights into the mechanism of countershading.
Collapse
Affiliation(s)
- Jiahao Chen
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Honggao Wang
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Shuting Wu
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Ao Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PRC
| | - Zhongkai Qiu
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Peng Huang
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jianan Y Qu
- Department of Electronic and Computer Engineering, The Hong Kong University of Science and Technology, Kowloon, China
| | - Jin Xu
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
13
|
Itoh T, Uehara M, Yura S, Wang JC, Fujii Y, Nakanishi A, Shimizu T, Hibi M. Foxp and Skor family proteins control differentiation of Purkinje cells from Ptf1a- and Neurog1-expressing progenitors in zebrafish. Development 2024; 151:dev202546. [PMID: 38456494 PMCID: PMC11057878 DOI: 10.1242/dev.202546] [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: 11/15/2023] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
Cerebellar neurons, such as GABAergic Purkinje cells (PCs), interneurons (INs) and glutamatergic granule cells (GCs) are differentiated from neural progenitors expressing proneural genes, including ptf1a, neurog1 and atoh1a/b/c. Studies in mammals previously suggested that these genes determine cerebellar neuron cell fate. However, our studies on ptf1a;neurog1 zebrafish mutants and lineage tracing of ptf1a-expressing progenitors have revealed that the ptf1a/neurog1-expressing progenitors can generate diverse cerebellar neurons, including PCs, INs and a subset of GCs in zebrafish. The precise mechanisms of how each cerebellar neuron type is specified remains elusive. We found that genes encoding the transcriptional regulators Foxp1b, Foxp4, Skor1b and Skor2, which are reportedly expressed in PCs, were absent in ptf1a;neurog1 mutants. foxp1b;foxp4 mutants showed a strong reduction in PCs, whereas skor1b;skor2 mutants completely lacked PCs, and displayed an increase in immature GCs. Misexpression of skor2 in GC progenitors expressing atoh1c suppressed GC fate. These data indicate that Foxp1b/4 and Skor1b/2 function as key transcriptional regulators in the initial step of PC differentiation from ptf1a/neurog1-expressing neural progenitors, and that Skor1b and Skor2 control PC differentiation by suppressing their differentiation into GCs.
Collapse
Affiliation(s)
- Tsubasa Itoh
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Mari Uehara
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Shinnosuke Yura
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Jui Chun Wang
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Yukimi Fujii
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Akiko Nakanishi
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Takashi Shimizu
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| | - Masahiko Hibi
- Department of Biological Science, Graduate School of Science, Nagoya University, Furo, Chikusa, Nagoya, Aichi 464-8602, Japan
| |
Collapse
|
14
|
Mulligan TS, Mumm JS. Selective Cell Ablation Using an Improved Prodrug-Converting Nitroreductase. Methods Mol Biol 2024; 2707:223-234. [PMID: 37668916 DOI: 10.1007/978-1-0716-3401-1_15] [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: 09/06/2023]
Abstract
Selective cell ablation is an invaluable tool to investigate the function of cell types, the regeneration of cells, and the modeling of diseases associated with cell loss. The nitroreductase (NTR)-mediated cell ablation system is a simple method enabling the elimination of targeted cells through the expression of a nitroreductase enzyme and the application of a prodrug (such as metronidazole). The prodrug is reduced to a cytotoxic product by nitroreductase, thereby leading to DNA damage-induced cell death. In species with elevated regenerative capacity such as zebrafish, removing the prodrug allows endogenous tissue to replace the lost cells. Herein, we describe a method for the use of a markedly improved nitroreductase enzyme for spatially and temporally controlled targeted cell ablation in the zebrafish. Recently, we identified an NTR variant (NTR 2.0) that achieves effective targeted cell ablation at concentrations of metronidazole well below those causing toxic side effects. NTR 2.0 thereby enables the ablation of "resistant" cell types and novel cell ablation paradigms. These advances simplify investigations of cell function, enable interrogations of the effects of chronic inflammation on regenerative processes and facilitate modeling of degenerative diseases associated with chronic cell loss. Techniques for transgenic nitroreductase expression and prodrug application are discussed.
Collapse
Affiliation(s)
- Timothy S Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
15
|
Tanday N, Tarasov AI, Moffett RC, Flatt PR, Irwin N. Pancreatic islet cell plasticity: Pathogenic or therapeutically exploitable? Diabetes Obes Metab 2024; 26:16-31. [PMID: 37845573 DOI: 10.1111/dom.15300] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 10/18/2023]
Abstract
The development of pancreatic islet endocrine cells is a tightly regulated process leading to the generation of distinct cell types harbouring different hormones in response to small changes in environmental stimuli. Cell differentiation is driven by transcription factors that are also critical for the maintenance of the mature islet cell phenotype. Alteration of the insulin-secreting β-cell transcription factor set by prolonged metabolic stress, associated with the pathogenesis of diabetes, obesity or pregnancy, results in the loss of β-cell identity through de- or transdifferentiation. Importantly, the glucose-lowering effects of approved and experimental antidiabetic agents, including glucagon-like peptide-1 mimetics, novel peptides and small molecules, have been associated with preventing or reversing β-cell dedifferentiation or promoting the transdifferentiation of non-β-cells towards an insulin-positive β-cell-like phenotype. Therefore, we review the manifestations of islet cell plasticity in various experimental settings and discuss the physiological and therapeutic sides of this phenomenon, focusing on strategies for preventing β-cell loss or generating new β-cells in diabetes. A better understanding of the molecular mechanisms underpinning islet cell plasticity is a prerequisite for more targeted therapies to help prevent β-cell decline in diabetes.
Collapse
Affiliation(s)
- Neil Tanday
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Andrei I Tarasov
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| | - R Charlotte Moffett
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| | - Peter R Flatt
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| | - Nigel Irwin
- Diabetes Research Centre, School of Biomedical Sciences, Ulster University, Coleraine, Northern Ireland
| |
Collapse
|
16
|
Rich MH, Sharrock AV, Mulligan TS, Matthews F, Brown AS, Lee-Harwood HR, Williams EM, Copp JN, Little RF, Francis JJB, Horvat CN, Stevenson LJ, Owen JG, Saxena MT, Mumm JS, Ackerley DF. A metagenomic library cloning strategy that promotes high-level expression of captured genes to enable efficient functional screening. Cell Chem Biol 2023; 30:1680-1691.e6. [PMID: 37898120 PMCID: PMC10842177 DOI: 10.1016/j.chembiol.2023.10.001] [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: 03/24/2023] [Revised: 06/17/2023] [Accepted: 10/02/2023] [Indexed: 10/30/2023]
Abstract
Functional screening of environmental DNA (eDNA) libraries is a potentially powerful approach to discover enzymatic "unknown unknowns", but is usually heavily biased toward the tiny subset of genes preferentially transcribed and translated by the screening strain. We have overcome this by preparing an eDNA library via partial digest with restriction enzyme FatI (cuts CATG), causing a substantial proportion of ATG start codons to be precisely aligned with strong plasmid-encoded promoter and ribosome-binding sequences. Whereas we were unable to select nitroreductases from standard metagenome libraries, our FatI strategy yielded 21 nitroreductases spanning eight different enzyme families, each conferring resistance to the nitro-antibiotic niclosamide and sensitivity to the nitro-prodrug metronidazole. We showed expression could be improved by co-expressing rare tRNAs and encoded proteins purified directly using an embedded His6-tag. In a transgenic zebrafish model of metronidazole-mediated targeted cell ablation, our lead MhqN-family nitroreductase proved ∼5-fold more effective than the canonical nitroreductase NfsB.
Collapse
Affiliation(s)
- Michelle H Rich
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Abigail V Sharrock
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Timothy S Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Frazer Matthews
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alistair S Brown
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Hannah R Lee-Harwood
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Elsie M Williams
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Janine N Copp
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Rory F Little
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Jenni J B Francis
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Claire N Horvat
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Luke J Stevenson
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Jeremy G Owen
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David F Ackerley
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand.
| |
Collapse
|
17
|
Emmerich K, Walker SL, Wang G, White DT, Ceisel A, Wang F, Teng Y, Chunawala Z, Graziano G, Nimmagadda S, Saxena MT, Qian J, Mumm JS. Transcriptomic comparison of two selective retinal cell ablation paradigms in zebrafish reveals shared and cell-specific regenerative responses. PLoS Genet 2023; 19:e1010905. [PMID: 37819938 PMCID: PMC10593236 DOI: 10.1371/journal.pgen.1010905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 10/23/2023] [Accepted: 08/07/2023] [Indexed: 10/13/2023] Open
Abstract
Retinal Müller glia (MG) can act as stem-like cells to generate new neurons in both zebrafish and mice. In zebrafish, retinal regeneration is innate and robust, resulting in the replacement of lost neurons and restoration of visual function. In mice, exogenous stimulation of MG is required to reveal a dormant and, to date, limited regenerative capacity. Zebrafish studies have been key in revealing factors that promote regenerative responses in the mammalian eye. Increased understanding of how the regenerative potential of MG is regulated in zebrafish may therefore aid efforts to promote retinal repair therapeutically. Developmental signaling pathways are known to coordinate regeneration following widespread retinal cell loss. In contrast, less is known about how regeneration is regulated in the context of retinal degenerative disease, i.e., following the loss of specific retinal cell types. To address this knowledge gap, we compared transcriptomic responses underlying regeneration following targeted loss of rod photoreceptors or bipolar cells. In total, 2,531 differentially expressed genes (DEGs) were identified, with the majority being paradigm specific, including during early MG activation phases, suggesting the nature of the injury/cell loss informs the regenerative process from initiation onward. For example, early modulation of Notch signaling was implicated in the rod but not bipolar cell ablation paradigm and components of JAK/STAT signaling were implicated in both paradigms. To examine candidate gene roles in rod cell regeneration, including several immune-related factors, CRISPR/Cas9 was used to create G0 mutant larvae (i.e., "crispants"). Rod cell regeneration was inhibited in stat3 crispants, while mutating stat5a/b, c7b and txn accelerated rod regeneration kinetics. These data support emerging evidence that discrete responses follow from selective retinal cell loss and that the immune system plays a key role in regulating "fate-biased" regenerative processes.
Collapse
Affiliation(s)
- Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Steven L. Walker
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Guohua Wang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - David T. White
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Anneliese Ceisel
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Fang Wang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| | - Zeeshaan Chunawala
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Gianna Graziano
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Saumya Nimmagadda
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Meera T. Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jiang Qian
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jeff S. Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, Maryland, United States of America
- McKusick-Nathans Institute of the Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
18
|
Matsuda H, Kubota Y. Zebrafish pancreatic β cell clusters undergo stepwise regeneration using Neurod1-expressing cells from different cell lineages. Cell Tissue Res 2023; 394:131-144. [PMID: 37474621 DOI: 10.1007/s00441-023-03805-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/30/2023] [Indexed: 07/22/2023]
Abstract
Pancreatic β cell clusters produce insulin and play a central role in glucose homeostasis. The regenerative capacity of mammalian β cells is limited and the loss of β cells causes diabetes. In contrast, zebrafish β cell clusters have a high regenerative capacity, making them an attractive model to study β cell cluster regeneration. How zebrafish β cell clusters regenerate, when the regeneration process is complete, and the identification of the cellular source of regeneration are fundamental questions that require investigation. Here, using larval and adult zebrafish, we demonstrate that pancreatic β cell clusters undergo a two-step regeneration process, regenerating functionality and then β cell numbers. Additionally, we found that all regenerating pancreatic β cells arose from Neurod1-expressing cells and that cells from different lineages contribute to both functional and β cell number recovery throughout their life. Furthermore, we found that during development and neogenesis, as well as regeneration, all β cells undergo Neurod1expression in zebrafish. Together, these results shed light on the fundamental cellular mechanisms underlying β cell cluster development, neogenesis, and regeneration.
Collapse
Affiliation(s)
- Hiroki Matsuda
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, Kusatsu, 525-8577, Japan.
- R&D division, Repli-tech Co., Ltd., Shibuya-ku, 150-0012, Japan.
| | - Yukihiko Kubota
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, 525-8577, Japan
| |
Collapse
|
19
|
Tucker TR, Knitter CA, Khoury DM, Eshghi S, Tran S, Sharrock AV, Wiles TJ, Ackerley DF, Mumm JS, Parsons MJ. An inducible model of chronic hyperglycemia. Dis Model Mech 2023; 16:dmm050215. [PMID: 37401381 PMCID: PMC10417516 DOI: 10.1242/dmm.050215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/28/2023] [Indexed: 07/05/2023] Open
Abstract
Transgene driven expression of Escherichia coli nitroreductase (NTR1.0) renders animal cells susceptible to the antibiotic metronidazole (MTZ). Many NTR1.0/MTZ ablation tools have been reported in zebrafish, which have significantly impacted regeneration studies. However, NTR1.0-based tools are not appropriate for modeling chronic cell loss as prolonged application of the required MTZ dose (10 mM) is deleterious to zebrafish health. We established that this dose corresponds to the median lethal dose (LD50) of MTZ in larval and adult zebrafish and that it induced intestinal pathology. NTR2.0 is a more active nitroreductase engineered from Vibrio vulnificus NfsB that requires substantially less MTZ to induce cell ablation. Here, we report on the generation of two new NTR2.0-based zebrafish lines in which acute β-cell ablation can be achieved without MTZ-associated intestinal pathology. For the first time, we were able to sustain β-cell loss and maintain elevated glucose levels (chronic hyperglycemia) in larvae and adults. Adult fish showed significant weight loss, consistent with the induction of a diabetic state, indicating that this paradigm will allow the modeling of diabetes and associated pathologies.
Collapse
Affiliation(s)
- Tori R. Tucker
- Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, Irvine, CA 92697, USA
| | - Courtney A. Knitter
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Natural Sciences II, Irvine, CA 92697, USA
| | - Deena M. Khoury
- Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, Irvine, CA 92697, USA
| | - Sheida Eshghi
- Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, Irvine, CA 92697, USA
| | - Sophia Tran
- Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, Irvine, CA 92697, USA
| | - Abigail V. Sharrock
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Travis J. Wiles
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Natural Sciences II, Irvine, CA 92697, USA
| | - David F. Ackerley
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Jeff S. Mumm
- Department of Ophthalmology, The Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Michael J. Parsons
- Department of Developmental and Cell Biology, University of California, Irvine, Natural Sciences II, Irvine, CA 92697, USA
| |
Collapse
|
20
|
Basile G, Vetere A, Hu J, Ijaduola O, Zhang Y, Liu KC, Eltony AM, De Jesus DF, Fukuda K, Doherty G, Leech CA, Chepurny OG, Holz GG, Yun SH, Andersson O, Choudhary A, Wagner BK, Kulkarni RN. Excess pancreatic elastase alters acinar-β cell communication by impairing the mechano-signaling and the PAR2 pathways. Cell Metab 2023; 35:1242-1260.e9. [PMID: 37339634 PMCID: PMC10834355 DOI: 10.1016/j.cmet.2023.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/21/2023] [Accepted: 05/17/2023] [Indexed: 06/22/2023]
Abstract
Type 1 (T1D) or type 2 diabetes (T2D) are caused by a deficit of functional insulin-producing β cells. Thus, the identification of β cell trophic agents could allow the development of therapeutic strategies to counteract diabetes. The discovery of SerpinB1, an elastase inhibitor that promotes human β cell growth, prompted us to hypothesize that pancreatic elastase (PE) regulates β cell viability. Here, we report that PE is up-regulated in acinar cells and in islets from T2D patients, and negatively impacts β cell viability. Using high-throughput screening assays, we identified telaprevir as a potent PE inhibitor that can increase human and rodent β cell viability in vitro and in vivo and improve glucose tolerance in insulin-resistant mice. Phospho-antibody microarrays and single-cell RNA sequencing analysis identified PAR2 and mechano-signaling pathways as potential mediators of PE. Taken together, our work highlights PE as a potential regulator of acinar-β cell crosstalk that acts to limit β cell viability, leading to T2D.
Collapse
Affiliation(s)
- Giorgio Basile
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Amedeo Vetere
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Jiang Hu
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Oluwaseun Ijaduola
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Yi Zhang
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Ka-Cheuk Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Amira M Eltony
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Dario F De Jesus
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Kazuki Fukuda
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Grace Doherty
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Colin A Leech
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA
| | - Oleg G Chepurny
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA
| | - George G Holz
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA; Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA
| | - Seok-Hyun Yun
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Amit Choudhary
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bridget K Wagner
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Rohit N Kulkarni
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA.
| |
Collapse
|
21
|
Lee MS, Han HJ, Choi TI, Lee KH, Baasankhuu A, Kim HT, Kim CH. IFT46 gene promoter-driven ciliopathy disease model in zebrafish. Front Cell Dev Biol 2023; 11:1200599. [PMID: 37363725 PMCID: PMC10285392 DOI: 10.3389/fcell.2023.1200599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Ciliopathies are human genetic disorders caused by abnormal formation and dysfunction of cellular cilia. Cilia are microtubule-based organelles that project into the extracellular space and transduce molecular and chemical signals from the extracellular environment or neighboring cells. Intraflagellar transport (IFT) proteins are required for the assembly and maintenance of cilia by transporting proteins along the axoneme which consists of complexes A and B. IFT46, a core IFT-B protein complex, is required for cilium formation and maintenance during vertebrate embryonic development. Here, we introduce transgenic zebrafish lines under the control of ciliated cell-specific IFT46 promoter to recapitulate human ciliopathy-like phenotypes. We generated a Tg(IFT46:GAL4-VP16) line to temporo-spatially control the expression of effectors including fluorescent reporters or nitroreductase based on the GAL4/UAS system, which expresses GAL4-VP16 chimeric transcription factors in most ciliated tissues during embryonic development. To analyze the function of IFT46-expressing ciliated cells during zebrafish development, we generated the Tg(IFT46:GAL4-VP16;UAS;nfsb-mCherry) line, a ciliated cell-specific injury model induced by nitroreductase (NTR)/metrodinazole (MTZ). Conditionally, controlled ablation of ciliated cells in transgenic animals exhibited ciliopathy-like phenotypes including cystic kidneys and pericardial and periorbital edema. Altogether, we established a zebrafish NTR/MTZ-mediated ciliated cell injury model that recapitulates ciliopathy-like phenotypes and may be a vertebrate animal model to further investigate the etiology and therapeutic approaches to human ciliopathies.
Collapse
Affiliation(s)
- Mi-Sun Lee
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
- Michigan Neuroscience Institute (MNI), University of Michigan, Ann Arbor, MI, United States
| | - Hye-Jeong Han
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Republic of Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Republic of Korea
| | - Tae-Ik Choi
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Kang-Han Lee
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Amartuvshin Baasankhuu
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Republic of Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Republic of Korea
| | - Hyun-Taek Kim
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan-Si, Republic of Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan-Si, Republic of Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
22
|
Coomer C, Naumova D, Talay M, Zolyomi B, Snell N, Sorkac A, Chanchu JM, Cheng J, Roman I, Li J, Robson D, Barnea G, Halpern ME. Transsynaptic labeling and transcriptional control of zebrafish neural circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535421. [PMID: 37066422 PMCID: PMC10103993 DOI: 10.1101/2023.04.03.535421] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Deciphering the connectome, the ensemble of synaptic connections that underlie brain function is a central goal of neuroscience research. The trans-Tango genetic approach, initially developed for anterograde transsynaptic tracing in Drosophila, can be used to map connections between presynaptic and postsynaptic partners and to drive gene expression in target neurons. Here, we describe the successful adaptation of trans-Tango to visualize neural connections in a living vertebrate nervous system, that of the zebrafish. Connections were validated between synaptic partners in the larval retina and brain. Results were corroborated by functional experiments in which optogenetic activation of retinal ganglion cells elicited responses in neurons of the optic tectum, as measured by trans-Tango-dependent expression of a genetically encoded calcium indicator. Transsynaptic signaling through trans-Tango reveals predicted as well as previously undescribed synaptic connections, providing a valuable in vivo tool to monitor and interrogate neural circuits over time.
Collapse
|
23
|
Boezio GLM, Zhao S, Gollin J, Priya R, Mansingh S, Guenther S, Fukuda N, Gunawan F, Stainier DYR. The developing epicardium regulates cardiac chamber morphogenesis by promoting cardiomyocyte growth. Dis Model Mech 2023; 16:dmm049571. [PMID: 36172839 PMCID: PMC9612869 DOI: 10.1242/dmm.049571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/13/2022] [Indexed: 11/20/2022] Open
Abstract
The epicardium, the outermost layer of the heart, is an important regulator of cardiac regeneration. However, a detailed understanding of the crosstalk between the epicardium and myocardium during development requires further investigation. Here, we generated three models of epicardial impairment in zebrafish by mutating the transcription factor genes tcf21 and wt1a, and ablating tcf21+ epicardial cells. Notably, all three epicardial impairment models exhibited smaller ventricles. We identified the initial cause of this phenotype as defective cardiomyocyte growth, resulting in reduced cell surface and volume. This failure of cardiomyocyte growth was followed by decreased proliferation and increased abluminal extrusion. By temporally manipulating its ablation, we show that the epicardium is required to support cardiomyocyte growth mainly during early cardiac morphogenesis. By transcriptomic profiling of sorted epicardial cells, we identified reduced expression of FGF and VEGF ligand genes in tcf21-/- hearts, and pharmacological inhibition of these signaling pathways in wild type partially recapitulated the ventricular growth defects. Taken together, these data reveal distinct roles of the epicardium during cardiac morphogenesis and signaling pathways underlying epicardial-myocardial crosstalk.
Collapse
Affiliation(s)
- Giulia L. M. Boezio
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Aulweg 130, 35392 Giessen, Germany
| | - Shengnan Zhao
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Josephine Gollin
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Rashmi Priya
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Aulweg 130, 35392 Giessen, Germany
| | - Shivani Mansingh
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Stefan Guenther
- Cardio-Pulmonary Institute, Aulweg 130, 35392 Giessen, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Nana Fukuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Felix Gunawan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Aulweg 130, 35392 Giessen, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- DZHK German Centre for Cardiovascular Research, Partner Site Rhine-Main, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute, Aulweg 130, 35392 Giessen, Germany
| |
Collapse
|
24
|
Kim J, Kim S, Choi WJ. Non-Invasive Monitoring of Cutaneous Wound Healing in Non-Diabetic and Diabetic Model of Adult Zebrafish Using OCT Angiography. Bioengineering (Basel) 2023; 10:bioengineering10050538. [PMID: 37237607 DOI: 10.3390/bioengineering10050538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
A diabetic wound presents a severe risk of infections and other complications because of its slow healing. Evaluating the pathophysiology during wound healing is imperative for wound care, necessitating a proper diabetic wound model and assay for monitoring. The adult zebrafish is a rapid and robust model for studying human cutaneous wound healing because of its fecundity and high similarities to human wound repair. OCTA as an assay can provide three-dimensional (3D) imaging of the tissue structure and vasculature in the epidermis, enabling monitoring of the pathophysiologic alterations in the zebrafish skin wound. We present a longitudinal study for assessing the cutaneous wound healing of the diabetic adult zebrafish model using OCTA, which is of importance for the diabetes research using the alternative animal models. We used non-diabetic (n = 9) and type 1 diabetes mellitus (DM) adult zebrafish models (n = 9). The full-thickness wound was generated on the fish skin, and the wound healing was monitored with OCTA for 15 days. The OCTA results demonstrated significant differences between diabetic and non-diabetic wound healing, involving delayed tissue remodeling and impaired angiogenesis for the diabetic wound, leading to slow wound recovery. The adult zebrafish model and OCTA technique may benefit long-term metabolic disease studies using zebrafish for drug development.
Collapse
Affiliation(s)
- Jaeyoung Kim
- Research Institute for Skin Image, Korea University College of Medicine, Seoul 08308, Republic of Korea
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
- Departments of Cancer Control Research and Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada
| | - Suhyun Kim
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Zebrafish Translational Medical Research Center, Korea University Ansan Hospital, Ansan 15355, Republic of Korea
| | - Woo June Choi
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
25
|
Kemmler CL, Moran HR, Murray BF, Scoresby A, Klem JR, Eckert RL, Lepovsky E, Bertho S, Nieuwenhuize S, Burger S, D'Agati G, Betz C, Puller AC, Felker A, Ditrychova K, Bötschi S, Affolter M, Rohner N, Lovely CB, Kwan KM, Burger A, Mosimann C. Next-generation plasmids for transgenesis in zebrafish and beyond. Development 2023; 150:dev201531. [PMID: 36975217 PMCID: PMC10263156 DOI: 10.1242/dev.201531] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Hannah R. Moran
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Brooke F. Murray
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Aaron Scoresby
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - John R. Klem
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rachel L. Eckert
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Elizabeth Lepovsky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Susan Nieuwenhuize
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Sibylle Burger
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Gianluca D'Agati
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Charles Betz
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Ann-Christin Puller
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Anastasia Felker
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Karolina Ditrychova
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Seraina Bötschi
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ben Lovely
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexa Burger
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
26
|
Diogo P, Martins G, Simão M, Marreiros A, Eufrásio AC, Cabrita E, Gavaia PJ. Type I Diabetes in Zebrafish Reduces Sperm Quality and Increases Insulin and Glucose Transporter Transcripts. Int J Mol Sci 2023; 24:ijms24087035. [PMID: 37108202 PMCID: PMC10138585 DOI: 10.3390/ijms24087035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Type I diabetes is a prominent human pathology with increasing incidence in the population; however, its cause is still unknown. This disease promotes detrimental effects on reproduction, such as lower sperm motility and DNA integrity. Hence, the investigation of the underlying mechanisms of this metabolic disturbance in reproduction and its transgenerational consequences is of the utmost importance. The zebrafish is a useful model for this research considering its high homology with human genes as well as its fast generation and regeneration abilities. Therefore, we aimed to investigate sperm quality and genes relevant to diabetes in the spermatozoa of Tg(ins:nfsb-mCherry) zebrafish, a model for type I diabetes. Diabetic Tg(ins:nfsb-mCherry) males showed significantly higher expression of transcripts for insulin a (insa) and glucose transporter (slc2a2) compared to controls. Sperm obtained from the same treatment group showed significantly lower sperm motility, plasma membrane viability, and DNA integrity compared to that from the control group. Upon sperm cryopreservation, sperm freezability was reduced, which could be a consequence of poor initial sperm quality. Altogether, the data showed similar detrimental effects related to type I diabetes in zebrafish spermatozoa at the cellular and molecular levels. Therefore, our study validates the zebrafish model for type I diabetes research in germ cells.
Collapse
Affiliation(s)
- Patrícia Diogo
- Faculty of Sciences and Technology (FCT), University of Algarve, 8005-139 Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Necton-Companhia Portuguesa de Culturas Marinhas S.A, Belamandil s/n, 8700-152 Olhão, Portugal
| | - Gil Martins
- Faculty of Sciences and Technology (FCT), University of Algarve, 8005-139 Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
| | - Márcio Simão
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
| | - Ana Marreiros
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
- Algarve Biomedical Center (ABC), University of Algarve, 8005-139 Faro, Portugal
| | - Ana Catarina Eufrásio
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), 4200-135 Porto, Portugal
| | - Elsa Cabrita
- Faculty of Sciences and Technology (FCT), University of Algarve, 8005-139 Faro, Portugal
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
| | - Paulo Jorge Gavaia
- Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, 8005-139 Faro, Portugal
| |
Collapse
|
27
|
Rich MH, Sharrock AV, Mulligan TS, Matthews F, Brown AS, Lee-Harwood HR, Williams EM, Copp JN, Little RF, Francis JJB, Horvat CN, Stevenson LJ, Owen JG, Saxena MT, Mumm JS, Ackerley DF. A metagenomic library cloning strategy that promotes high-level expression of captured genes to enable efficient functional screening. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.534183. [PMID: 36993673 PMCID: PMC10055417 DOI: 10.1101/2023.03.24.534183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Functional screening of environmental DNA (eDNA) libraries is a potentially powerful approach to discover enzymatic "unknown unknowns", but is usually heavily biased toward the tiny subset of genes preferentially transcribed and translated by the screening strain. We have overcome this by preparing an eDNA library via partial digest with restriction enzyme FatI (cuts CATG), causing a substantial proportion of ATG start codons to be precisely aligned with strong plasmid-encoded promoter and ribosome-binding sequences. Whereas we were unable to select nitroreductases from standard metagenome libraries, our FatI strategy yielded 21 nitroreductases spanning eight different enzyme families, each conferring resistance to the nitro-antibiotic niclosamide and sensitivity to the nitro-prodrug metronidazole. We showed expression could be improved by co-expressing rare tRNAs and encoded proteins purified directly using an embedded His6-tag. In a transgenic zebrafish model of metronidazole-mediated targeted cell ablation, our lead MhqN-family nitroreductase proved ~5-fold more effective than the canonical nitroreductase NfsB.
Collapse
Affiliation(s)
- Michelle H Rich
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Abigail V Sharrock
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Timothy S Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Frazer Matthews
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alistair S Brown
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Hannah R Lee-Harwood
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Elsie M Williams
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Current address: Burnet Institute, Melbourne, Victoria 3004, Australia
| | - Janine N Copp
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Current addresses: Michael Smith Laboratories, University of British Columbia, Vancouver BC V6T 1Z4, Canada; Abcellera Biologics Inc, Vancouver BC V5Y 0A1, Canada
| | - Rory F Little
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Current address: Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany
| | - Jenni JB Francis
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Claire N Horvat
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Current address: Teva Pharmaceuticals, Sydney, New South Wales 2113, Australia
| | - Luke J Stevenson
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Jeremy G Owen
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David F Ackerley
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
28
|
Research Progress on the Construction and Application of a Diabetic Zebrafish Model. Int J Mol Sci 2023; 24:ijms24065195. [PMID: 36982274 PMCID: PMC10048833 DOI: 10.3390/ijms24065195] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Diabetes is a metabolic disease characterized by high blood glucose levels. With economic development and lifestyle changes, the prevalence of diabetes is increasing yearly. Thus, it has become an increasingly serious public health problem in countries around the world. The etiology of diabetes is complex, and its pathogenic mechanisms are not completely clear. The use of diabetic animal models is helpful in the study of the pathogenesis of diabetes and the development of drugs. The emerging vertebrate model of zebrafish has many advantages, such as its small size, large number of eggs, short growth cycle, simple cultivation of adult fish, and effective improvement of experimental efficiency. Thus, this model is highly suitable for research as an animal model of diabetes. This review not only summarizes the advantages of zebrafish as a diabetes model, but also summarizes the construction methods and challenges of zebrafish models of type 1 diabetes, type 2 diabetes, and diabetes complications. This study provides valuable reference information for further study of the pathological mechanisms of diabetes and the research and development of new related therapeutic drugs.
Collapse
|
29
|
Sargent S, Brennan A, Clark JK. Regenerative potential and limitations in a zebrafish model of hyperglycemia-induced nerve degeneration. Dev Dyn 2023. [PMID: 36879394 DOI: 10.1002/dvdy.583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/04/2023] [Accepted: 02/25/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Previous work from our lab has described a model of motor nerve degeneration in hyperglycemic zebrafish larvae which resembles mammalian models of diabetic peripheral neuropathy (DPN). Here, we optimized the hyperglycemic-induction protocol, characterized deficits in nerve structure and behavioral function, and then examined the regenerative potential following recovery from the hyperglycemic state. RESULTS In agreement with our previous work, hyperglycemia induced motor nerve degeneration and behavioral deficits. However, the optimized protocol initiated disruption of tight junctions within the blood-nerve barrier, a phenotype apparent in mammalian models of DPN. Following a 10-day recovery period, regeneration of motor nerve components was apparent, but behavioral deficits persisted. We next examined the effect of hyperglycemia on the musculoskeletal system and found subtle deficits in muscle that resolved following recovery, and robust deficits in the skeletal system which persisted following recovery. CONCLUSION Here we optimized our previous model of hyperglycemia-induced motor nerve degeneration to more closely align with that observed in mammalian models and then characterized the regenerative potential following recovery from hyperglycemia. Notably, we observed striking impairments to skeletal development, which underscores the global impact hyperglycemia has across systems, and provides a framework for elucidating molecular mechanisms responsible for regenerative events moving forward.
Collapse
Affiliation(s)
- Sheridan Sargent
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland, USA
| | - Anna Brennan
- Department of Biological Sciences, Salisbury University, Salisbury, Maryland, USA
| | | |
Collapse
|
30
|
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
|
31
|
Sudhakaran G, Rajesh R, Guru A, Haridevamuthu B, Murugan R, Bhuvanesh N, Wadaan MA, Mahboob S, Juliet A, Gopinath P, Arockiaraj J. Deacetylated nimbin analog N2 fortifies alloxan-induced pancreatic β-cell damage in insulin-resistant zebrafish larvae by upregulating phosphoenolpyruvate carboxykinase (PEPCK) and insulin levels. Toxicol Appl Pharmacol 2022; 454:116229. [PMID: 36089001 DOI: 10.1016/j.taap.2022.116229] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022]
Abstract
This study aims to evaluate the protective behaviour of N2, a semi-natural analog of nimbin, for its anti-diabetic efficacy against alloxan-induced oxidative damage and β-cell dysfunction in in-vivo zebrafish larvae. A 500 μM of alloxan was exposed to zebrafish larvae for 24 h to induce oxidative stress in the pancreatic β-cells and co-exposed with N2 to study the protection of N2 by inhibiting ROS by DCFH-DA, DHE and NDA staining along with Cellular damage, apoptosis and lipid peroxidation. The zebrafish was further exposed to 500 μM alloxan for 72 h to induce β-cell destruction along with depleted glucose uptake and co-exposed to N2 to study the protective mechanism. Glucose levels were estimated, and PCR was used to verify the mRNA expression of phosphoenolpyruvate carboxykinase (PEPCK) and insulin. Alloxan induced (24 h) oxidative stress in the pancreatic β-cells in which N2's co-exposure inhibited ROS by eliminating O-₂ radicals and restoring the glutathione levels, thus preventing cellular damage and lipid peroxidation. The zebrafish exposed to 500 μM alloxan for 72 h was observed with β-cell destruction along with depleted glucose uptake when stained with 2NBDG, wherein N2 was able to protect the pancreatic β-cells from oxidative damage, promoted high glucose uptake and reduced glucose levels. N2 stimulated insulin production and downregulated PEPCK by inhibiting gluconeogenesis, attenuating post-prandial hyperglycemia. N2 may contribute to anti-oxidant protection against alloxan-induced β-cell damage and anti-hyperglycemic activity, restoring insulin function and suppressing PEPCK expression.
Collapse
Affiliation(s)
- Gokul Sudhakaran
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Ravi Rajesh
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Ajay Guru
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - B Haridevamuthu
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Raghul Murugan
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India
| | - Nattamai Bhuvanesh
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA
| | - Mohammad Ahmad Wadaan
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Shalid Mahboob
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Annie Juliet
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, University Station A4800, Austin, TX 78712, USA
| | - Pushparathinam Gopinath
- Department of Chemistry, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Department of Biotechnology, College of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, 603 203 Chennai, Tamil Nadu, India.
| |
Collapse
|
32
|
Livne H, Avital T, Ruppo S, Harazi A, Mitrani-Rosenbaum S, Daya A. Generation and characterization of a novel gne Knockout Model in Zebrafish. Front Cell Dev Biol 2022; 10:976111. [PMID: 36353515 PMCID: PMC9637792 DOI: 10.3389/fcell.2022.976111] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/14/2022] [Indexed: 12/04/2022] Open
Abstract
GNE Myopathy is a rare, recessively inherited neuromuscular worldwide disorder, caused by a spectrum of bi-allelic mutations in the human GNE gene. GNE encodes a bi-functional enzyme responsible for the rate-limiting step of sialic acid biosynthesis pathway. However, the process in which GNE mutations lead to the development of a muscle pathology is not clear yet. Cellular and mouse models for GNE Myopathy established to date have not been informative. Further, additional GNE functions in muscle have been hypothesized. In these studies, we aimed to investigate gne functions using zebrafish genetic and transgenic models, and characterized them using macroscopic, microscopic, and molecular approaches. We first established transgenic zebrafish lineages expressing the human GNE cDNA carrying the M743T mutation, driven by the zebrafish gne promoter. These fish developed entirely normally. Then, we generated a gne knocked-out (KO) fish using the CRISPR/Cas9 methodology. These fish died 8–10 days post-fertilization (dpf), but a phenotype appeared less than 24 h before death and included progressive body axis curving, deflation of the swim bladder and decreasing movement and heart rate. However, muscle histology uncovered severe defects, already at 5 dpf, with compromised fiber organization. Sialic acid supplementation did not rescue the larvae from this phenotype nor prolonged their lifespan. To have deeper insights into the potential functions of gne in zebrafish, RNA sequencing was performed at 3 time points (3, 5, and 7 dpf). Genotype clustering was progressive, with only 5 genes differentially expressed in gne KO compared to gne WT siblings at 3 dpf. Enrichment analyses of the primary processes affected by the lack of gne also at 5 and 7 dpf point to the involvement of cell cycle and DNA damage/repair processes in the gne KO zebrafish. Thus, we have established a gne KO zebrafish lineage and obtained new insights into gne functions. This is the only model where GNE can be related to clear muscle defects, thus the only animal model relevant to GNE Myopathy to date. Further elucidation of gne precise mechanism-of-action in these processes could be relevant to GNE Myopathy and allow the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Hagay Livne
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tom Avital
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
| | - Shmuel Ruppo
- Info-CORE, Bioinformatics Unit of the I-CORE, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Avi Harazi
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Stella Mitrani-Rosenbaum
- Goldyne Savad Institute of Gene Therapy, Hadassah Medical Center, The Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Alon Daya
- Faculty of Marine Sciences, Ruppin Academic Center, Michmoret, Israel
- *Correspondence: Alon Daya,
| |
Collapse
|
33
|
Fitzpatrick MJ, Kerschensteiner D. Homeostatic plasticity in the retina. Prog Retin Eye Res 2022; 94:101131. [PMID: 36244950 DOI: 10.1016/j.preteyeres.2022.101131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 02/07/2023]
Abstract
Vision begins in the retina, whose intricate neural circuits extract salient features of the environment from the light entering our eyes. Neurodegenerative diseases of the retina (e.g., inherited retinal degenerations, age-related macular degeneration, and glaucoma) impair vision and cause blindness in a growing number of people worldwide. Increasing evidence indicates that homeostatic plasticity (i.e., the drive of a neural system to stabilize its function) can, in principle, preserve retinal function in the face of major perturbations, including neurodegeneration. Here, we review the circumstances and events that trigger homeostatic plasticity in the retina during development, sensory experience, and disease. We discuss the diverse mechanisms that cooperate to compensate and the set points and outcomes that homeostatic retinal plasticity stabilizes. Finally, we summarize the opportunities and challenges for unlocking the therapeutic potential of homeostatic plasticity. Homeostatic plasticity is fundamental to understanding retinal development and function and could be an important tool in the fight to preserve and restore vision.
Collapse
|
34
|
Duque M, Amorim JP, Bessa J. Ptf1a function and transcriptional cis-regulation, a cornerstone in vertebrate pancreas development. FEBS J 2022; 289:5121-5136. [PMID: 34125483 PMCID: PMC9545688 DOI: 10.1111/febs.16075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/23/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022]
Abstract
Vertebrate pancreas organogenesis is a stepwise process regulated by a complex network of signaling and transcriptional events, progressively steering the early endoderm toward pancreatic fate. Many crucial players of this process have been identified, including signaling pathways, cis-regulatory elements, and transcription factors (TFs). Pancreas-associated transcription factor 1a (PTF1A) is one such TF, crucial for pancreas development. PTF1A mutations result in dramatic pancreatic phenotypes associated with severe complications, such as neonatal diabetes and impaired food digestion due to exocrine pancreatic insufficiency. Here, we present a brief overview of vertebrate pancreas development, centered on Ptf1a function and transcriptional regulation, covering similarities and divergences in three broadly studied organisms: human, mouse and zebrafish.
Collapse
Affiliation(s)
- Marta Duque
- Instituto de Biologia Molecular e Celular (IBMC)Universidade do PortoPortugal
- Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortugal
- Doctoral program in Molecular and Cell Biology (MCbiology)Instituto de Ciências Biomédicas Abel Salazar (ICBAS)Universidade do PortoPortugal
| | - João Pedro Amorim
- Instituto de Biologia Molecular e Celular (IBMC)Universidade do PortoPortugal
- Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortugal
- Doctoral program in Molecular and Cell Biology (MCbiology)Instituto de Ciências Biomédicas Abel Salazar (ICBAS)Universidade do PortoPortugal
| | - José Bessa
- Instituto de Biologia Molecular e Celular (IBMC)Universidade do PortoPortugal
- Instituto de Investigação e Inovação em Saúde (i3S)Universidade do PortoPortugal
| |
Collapse
|
35
|
Macrophages and neutrophils are necessary for ER stress-induced β cell loss. Cell Rep 2022; 40:111255. [PMID: 36001973 PMCID: PMC9444341 DOI: 10.1016/j.celrep.2022.111255] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/09/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Persistent endoplasmic reticulum (ER) stress induces islet inflammation and β cell loss. How islet inflammation contributes to β cell loss remains uncertain. We have reported previously that chronic overnutrition-induced ER stress in β cells causes Ripk3-mediated islet inflammation, macrophage recruitment, and a reduction of β cell numbers in a zebrafish model. We show here that β cell loss results from the intricate communications among β cells, macrophages, and neutrophils. Macrophage-derived Tnfa induces cxcl8a in β cells. Cxcl8a, in turn, attracts neutrophils to macrophage-contacted “hotspots” where β cell loss occurs. We also show potentiation of chemokine expression in stressed mammalian β cells by macrophage-derived TNFA. In Akita and db/db mice, there is an increase in CXCL15-positive β cells and intra-islet neutrophils. Blocking neutrophil recruitment in Akita mice preserves β cell mass and slows diabetes progression. These results reveal an important role of neutrophils in persistent ER stress-induced β cell loss. Yang et al. show a pivotal role of communications among β cells, macrophages, and neutrophils in chronic overnutrition-induced loss of pancreatic β cells in a diabetes-prone zebrafish model.
Collapse
|
36
|
Bruton FA, Kaveh A, Ross-Stewart KM, Matrone G, Oremek MEM, Solomonidis EG, Tucker CS, Mullins JJ, Lucas CD, Brittan M, Taylor JM, Rossi AG, Denvir MA. Macrophages trigger cardiomyocyte proliferation by increasing epicardial vegfaa expression during larval zebrafish heart regeneration. Dev Cell 2022; 57:1512-1528.e5. [PMID: 35688158 PMCID: PMC9616726 DOI: 10.1016/j.devcel.2022.05.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/11/2022] [Accepted: 05/18/2022] [Indexed: 12/01/2022]
Abstract
Cardiac injury leads to the loss of cardiomyocytes, which are rapidly replaced by the proliferation of the surviving cells in zebrafish, but not in mammals. In both the regenerative zebrafish and non-regenerative mammals, cardiac injury induces a sustained macrophage response. Macrophages are required for cardiomyocyte proliferation during zebrafish cardiac regeneration, but the mechanisms whereby macrophages facilitate this crucial process are fundamentally unknown. Using heartbeat-synchronized live imaging, RNA sequencing, and macrophage-null genotypes in the larval zebrafish cardiac injury model, we characterize macrophage function and reveal that these cells activate the epicardium, inducing cardiomyocyte proliferation. Mechanistically, macrophages are specifically recruited to the epicardial-myocardial niche, triggering the expansion of the epicardium, which upregulates vegfaa expression to induce cardiomyocyte proliferation. Our data suggest that epicardial Vegfaa augments a developmental cardiac growth pathway via increased endocardial notch signaling. The identification of this macrophage-dependent mechanism of cardiac regeneration highlights immunomodulation as a potential strategy for enhancing mammalian cardiac repair. Heart regeneration in larval zebrafish is characterized in detail Macrophage ablation blocks cardiomyocyte proliferation after cardiac injury Macrophages synapse with epicardial cells and promote their proliferation Epicardial Vegfaa drives cardiomyocyte proliferation during cardiac regeneration
Collapse
Affiliation(s)
- Finnius A Bruton
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| | - Aryan Kaveh
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK; Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine M Ross-Stewart
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Gianfranco Matrone
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Magdalena E M Oremek
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Emmanouil G Solomonidis
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Carl S Tucker
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - John J Mullins
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Christopher D Lucas
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | - Adriano G Rossi
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Martin A Denvir
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
37
|
Hall CJ, Astin JW, Mumm JS, Ackerley DF. A New Transgenic Line for Rapid and Complete Neutrophil Ablation. Zebrafish 2022; 19:109-113. [PMID: 35617702 DOI: 10.1089/zeb.2022.0020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Zebrafish lines expressing nitroreductase (NTR) in specific cell compartments, which sensitizes those cells to metronidazole (MTZ)-mediated ablation, have proven extremely useful for studying tissue regeneration and investigating cell function. In contrast to many cells, neutrophils are comparatively resistant to the NTR/MTZ targeted ablation strategy. Recently, a rationally engineered variant of NTR (NTR 2.0) has been described that exhibits greatly improved MTZ-mediated ablation efficacy in zebrafish. We show that a transgenic line with neutrophil-restricted expression of NTR 2.0 demonstrates complete neutrophil ablation, with an MTZ dose 100-fold less than current treatment regimens, and with treatment durations as short as 5 h.
Collapse
Affiliation(s)
- Christopher J Hall
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jonathan W Astin
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jeff S Mumm
- Department of Ophthalmology, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Human Genetics, Johns Hopkins University, Baltimore, Maryland, USA
| | - David F Ackerley
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
38
|
Corradi L, Bruzzone M, Maschio MD, Sawamiphak S, Filosa A. Hypothalamic Galanin-producing neurons regulate stress in zebrafish through a peptidergic, self-inhibitory loop. Curr Biol 2022; 32:1497-1510.e5. [PMID: 35219430 DOI: 10.1016/j.cub.2022.02.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/10/2022] [Accepted: 02/02/2022] [Indexed: 12/24/2022]
Abstract
Animals possess neuronal circuits inducing stress to avoid or cope with threats present in their surroundings, for instance, by promoting behaviors, such as avoidance and escape. However, mechanisms must exist to tightly control responses to stressors, since overactivation of stress circuits is deleterious for the wellbeing of an organism. The underlying neuronal dynamics responsible for controlling behavioral responses to stress have remained unclear. Here, we describe a neuronal circuit in the hypothalamus of zebrafish larvae that inhibits stress-related behaviors and prevents excessive activation of the neuroendocrine pathway hypothalamic-pituitary-interrenal axis. Central components of this circuit are neurons secreting the neuropeptide Galanin, as ablation of these neurons led to abnormally high levels of stress. Surprisingly, we found that Galanin has a self-inhibitory action on Galanin-producing neurons. Our results suggest that hypothalamic Galanin-producing neurons play an important role in fine-tuning stress responses by preventing potentially harmful overactivation of stress-regulating circuits.
Collapse
Affiliation(s)
- Laura Corradi
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Freie Universität Berlin, Institute for Biology, Berlin, Germany
| | - Matteo Bruzzone
- Padova Neuroscience Center, Università degli Studi di Padova, Padua, Italy
| | - Marco Dal Maschio
- Padova Neuroscience Center, Università degli Studi di Padova, Padua, Italy; Department of Biomedical Sciences, Università degli Studi di Padova, Padua, Italy
| | - Suphansa Sawamiphak
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Alessandro Filosa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
| |
Collapse
|
39
|
Lin T, Wang S, Munker S, Jung K, Macías-Rodríguez RU, Ruiz-Margáin A, Schierwagen R, Liu H, Shao C, Fan C, Feng R, Yuan X, Wang S, Wandrer F, Meyer C, Wimmer R, Liebe R, Kroll J, Zhang L, Schiergens T, Ten Dijke P, Teufel A, Marx A, Mertens PR, Wang H, Ebert MPA, Bantel H, N De Toni E, Trebicka J, Dooley S, Shin D, Ding H, Weng HL. Follistatin-controlled activin-HNF4α-coagulation factor axis in liver progenitor cells determines outcome of acute liver failure. Hepatology 2022; 75:322-337. [PMID: 34435364 DOI: 10.1002/hep.32119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/01/2021] [Accepted: 08/15/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS In patients with acute liver failure (ALF) who suffer from massive hepatocyte loss, liver progenitor cells (LPCs) take over key hepatocyte functions, which ultimately determines survival. This study investigated how the expression of hepatocyte nuclear factor 4α (HNF4α), its regulators, and targets in LPCs determines clinical outcome of patients with ALF. APPROACH AND RESULTS Clinicopathological associations were scrutinized in 19 patients with ALF (9 recovered and 10 receiving liver transplantation). Regulatory mechanisms between follistatin, activin, HNF4α, and coagulation factor expression in LPC were investigated in vitro and in metronidazole-treated zebrafish. A prospective clinical study followed up 186 patients with cirrhosis for 80 months to observe the relevance of follistatin levels in prevalence and mortality of acute-on-chronic liver failure. Recovered patients with ALF robustly express HNF4α in either LPCs or remaining hepatocytes. As in hepatocytes, HNF4α controls the expression of coagulation factors by binding to their promoters in LPC. HNF4α expression in LPCs requires the forkhead box protein H1-Sma and Mad homolog 2/3/4 transcription factor complex, which is promoted by the TGF-β superfamily member activin. Activin signaling in LPCs is negatively regulated by follistatin, a hepatocyte-derived hormone controlled by insulin and glucagon. In contrast to patients requiring liver transplantation, recovered patients demonstrate a normal activin/follistatin ratio, robust abundance of the activin effectors phosphorylated Sma and Mad homolog 2 and HNF4α in LPCs, leading to significantly improved coagulation function. A follow-up study indicated that serum follistatin levels could predict the incidence and mortality of acute-on-chronic liver failure. CONCLUSIONS These results highlight a crucial role of the follistatin-controlled activin-HNF4α-coagulation axis in determining the clinical outcome of massive hepatocyte loss-induced ALF. The effects of insulin and glucagon on follistatin suggest a key role of the systemic metabolic state in ALF.
Collapse
Affiliation(s)
- Tao Lin
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Shanshan Wang
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- Beijing Institute of HepatologyBeijing You'an HospitalCapital Medical UniversityBeijingChina
| | - Stefan Munker
- Department of Medicine IIUniversity Hospital, Campus Großhadern, LMU MunichMunichGermany
| | - Kyounghwa Jung
- Department of Developmental BiologyMcGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Ricardo U Macías-Rodríguez
- Department of GastroenterologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico cityMexico
| | - Astrid Ruiz-Margáin
- Department of GastroenterologyInstituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránMexico cityMexico
| | - Robert Schierwagen
- Translational Hepatology, Medical Department IFrankfurt University HospitalFrankfurtGermany
| | - Hui Liu
- Department of PathologyBeijing You'an HospitalAffiliated with Capital Medical UniversityBeijingChina
| | - Chen Shao
- Department of PathologyBeijing You'an HospitalAffiliated with Capital Medical UniversityBeijingChina
| | - Chunlei Fan
- Department of Gastroenterology and HepatologyBeijing You'an HospitalAffiliated with Capital Medical UniversityBeijingChina
| | - Rilu Feng
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Xiaodong Yuan
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Sai Wang
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Franziska Wandrer
- Department of Gastroenterology, Hepatology and EndocrinologyHannover Medical SchoolHannoverGermany
| | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Ralf Wimmer
- Department of Medicine IIUniversity Hospital, Campus Großhadern, LMU MunichMunichGermany
| | - Roman Liebe
- Clinic of Gastroenterology, Hepatology and Infectious DiseasesHeinrich Heine UniversityDüsseldorfGermany
- Department of Medicine IISaarland University Medical CenterSaarland UniversityHomburgGermany
| | - Jens Kroll
- Vascular Biology and Tumor AngiogenesisEuropean Center for AngioscienceMedical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Long Zhang
- Life Sciences Institute and Innovation Center for Cell Signaling NetworkHangzhouChina
| | - Tobias Schiergens
- Department of General, Visceral, Transplantation, Vascular and Thoracic SurgeryUniversity HospitalLMU MunichMunichGermany
| | - Peter Ten Dijke
- Oncode Institute and Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Andreas Teufel
- Division of Hepatology, Division of Clinical Bioinformatics, Department of Medicine II, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- Clinical Cooperation Unit Healthy MetabolismCenter for Preventive Medicine and Digital Health Baden-WürttembergMedical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Alexander Marx
- Institute of PathologyUniversity Medical Center MannheimHeidelberg UniversityMannheimGermany
| | - Peter R Mertens
- Clinic of Nephrology and Hypertension, Diabetes and EndocrinologyOtto-von-Guericke-UniversityMagdeburgGermany
| | - Hua Wang
- Department of Oncologythe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Disease Laboratory of Anhui ProvinceHefeiChina
| | - Matthias P A Ebert
- Mannheim Institute for Innate ImmunoscienceMannheimGermany
- Clinical Cooperation Unit Healthy MetabolismCenter of Preventive Medicine and Digital HealthMedical Faculty MannheimHeidelberg UniversityMannheimGermany
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology and EndocrinologyHannover Medical SchoolHannoverGermany
| | - Enrico N De Toni
- Department of Medicine IIUniversity Hospital, Campus Großhadern, LMU MunichMunichGermany
| | - Jonel Trebicka
- Translational Hepatology, Medical Department IFrankfurt University HospitalFrankfurtGermany
- European Foundation for Study of Chronic Liver FailureBarcelonaSpain
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| | - Donghun Shin
- Department of Developmental BiologyMcGowan Institute for Regenerative MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Huiguo Ding
- Department of Gastroenterology and HepatologyBeijing You'an HospitalAffiliated with Capital Medical UniversityBeijingChina
| | - Hong-Lei Weng
- Department of Medicine II, Section Molecular Hepatology, University Medical Center Mannheim, Medical Faculty MannheimHeidelberg UniversityMannheimGermany
| |
Collapse
|
40
|
Chowdhury K, Lin S, Lai SL. Comparative Study in Zebrafish and Medaka Unravels the Mechanisms of Tissue Regeneration. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.783818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tissue regeneration has been in the spotlight of research for its fascinating nature and potential applications in human diseases. The trait of regenerative capacity occurs diversely across species and tissue contexts, while it seems to decline over evolution. Organisms with variable regenerative capacity are usually distinct in phylogeny, anatomy, and physiology. This phenomenon hinders the feasibility of studying tissue regeneration by directly comparing regenerative with non-regenerative animals, such as zebrafish (Danio rerio) and mice (Mus musculus). Medaka (Oryzias latipes) is a fish model with a complete reference genome and shares a common ancestor with zebrafish approximately 110–200 million years ago (compared to 650 million years with mice). Medaka shares similar features with zebrafish, including size, diet, organ system, gross anatomy, and living environment. However, while zebrafish regenerate almost every organ upon experimental injury, medaka shows uneven regenerative capacity. Their common and distinct biological features make them a unique platform for reciprocal analyses to understand the mechanisms of tissue regeneration. Here we summarize current knowledge about tissue regeneration in these fish models in terms of injured tissues, repairing mechanisms, available materials, and established technologies. We further highlight the concept of inter-species and inter-organ comparisons, which may reveal mechanistic insights and hint at therapeutic strategies for human diseases.
Collapse
|
41
|
Sharrock AV, Mulligan TS, Hall KR, Williams EM, White DT, Zhang L, Emmerich K, Matthews F, Nimmagadda S, Washington S, Le KD, Meir-Levi D, Cox OL, Saxena MT, Calof AL, Lopez-Burks ME, Lander AD, Ding D, Ji H, Ackerley DF, Mumm JS. NTR 2.0: a rationally engineered prodrug-converting enzyme with substantially enhanced efficacy for targeted cell ablation. Nat Methods 2022; 19:205-215. [PMID: 35132245 PMCID: PMC8851868 DOI: 10.1038/s41592-021-01364-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/29/2021] [Indexed: 11/12/2022]
Abstract
Transgenic expression of bacterial nitroreductase (NTR) enzymes sensitizes eukaryotic cells to prodrugs such as metronidazole (MTZ), enabling selective cell-ablation paradigms that have expanded studies of cell function and regeneration in vertebrates. However, first-generation NTRs required confoundingly toxic prodrug treatments to achieve effective cell ablation, and some cell types have proven resistant. Here we used rational engineering and cross-species screening to develop an NTR variant, NTR 2.0, which exhibits ~100-fold improvement in MTZ-mediated cell-specific ablation efficacy, eliminating the need for near-toxic prodrug treatment regimens. NTR 2.0 therefore enables sustained cell-loss paradigms and ablation of previously resistant cell types. These properties permit enhanced interrogations of cell function, extended challenges to the regenerative capacities of discrete stem cell niches, and novel modeling of chronic degenerative diseases. Accordingly, we have created a series of bipartite transgenic reporter/effector resources to facilitate dissemination of NTR 2.0 to the research community.
Collapse
Affiliation(s)
- Abigail V Sharrock
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Timothy S Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kelsi R Hall
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Elsie M Williams
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - David T White
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Liyun Zhang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Frazer Matthews
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Saumya Nimmagadda
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Selena Washington
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Katherine D Le
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Danielle Meir-Levi
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Olivia L Cox
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA
- Luminomics, Baltimore, MD, USA
| | - Anne L Calof
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Martha E Lopez-Burks
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Arthur D Lander
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
| | - Ding Ding
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David F Ackerley
- School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand.
- Centre for Biodiscovery and Maurice Wilkins Centre for Molecular Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.
- Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
42
|
Singh SP, Chawla P, Hnatiuk A, Kamel M, Silva LD, Spanjaard B, Eski SE, Janjuha S, Olivares-Chauvet P, Kayisoglu O, Rost F, Bläsche J, Kränkel A, Petzold A, Kurth T, Reinhardt S, Junker JP, Ninov N. A single-cell atlas of de novo β-cell regeneration reveals the contribution of hybrid β/δ-cells to diabetes recovery in zebrafish. Development 2022; 149:274140. [PMID: 35088828 DOI: 10.1242/dev.199853] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022]
Abstract
Regeneration-competent species possess the ability to reverse the progression of severe diseases by restoring the function of the damaged tissue. However, the cellular dynamics underlying this capability remain unexplored. Here, we have used single-cell transcriptomics to map de novo β-cell regeneration during induction and recovery from diabetes in zebrafish. We show that the zebrafish has evolved two distinct types of somatostatin-producing δ-cells, which we term δ1- and δ2-cells. Moreover, we characterize a small population of glucose-responsive islet cells, which share the hormones and fate-determinants of both β- and δ1-cells. The transcriptomic analysis of β-cell regeneration reveals that β/δ hybrid cells provide a prominent source of insulin expression during diabetes recovery. Using in vivo calcium imaging and cell tracking, we further show that the hybrid cells form de novo and acquire glucose-responsiveness in the course of regeneration. The overexpression of dkk3, a gene enriched in hybrid cells, increases their formation in the absence of β-cell injury. Finally, interspecies comparison shows that plastic δ1-cells are partially related to PP cells in the human pancreas. Our work provides an atlas of β-cell regeneration and indicates that the rapid formation of glucose-responsive hybrid cells contributes to the resolution of diabetes in zebrafish.
Collapse
Affiliation(s)
- Sumeet Pal Singh
- IRIBHM, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Prateek Chawla
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Alisa Hnatiuk
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Margrit Kamel
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Luis Delgadillo Silva
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Bastiaan Spanjaard
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Sema Elif Eski
- IRIBHM, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Sharan Janjuha
- Institute of Pharmacology and Toxicology, University of Zurich, CH-8057 Zurich, Switzerland
| | - Pedro Olivares-Chauvet
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Oezge Kayisoglu
- The Julius Maximilian University of Wurzburg, 97070 Wurzburg, Germany
| | - Fabian Rost
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany.,DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Juliane Bläsche
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Annekathrin Kränkel
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Andreas Petzold
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Thomas Kurth
- TUD, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform, EM-Facility, Technische Universitaät Dresden, 01307 Dresden, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, DFG NGS Competence Center, c/o Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Jan Philipp Junker
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115 Berlin, Germany
| | - Nikolay Ninov
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
43
|
MNK2 deficiency potentiates β-cell regeneration via translational regulation. Nat Chem Biol 2022; 18:942-953. [PMID: 35697798 PMCID: PMC7613404 DOI: 10.1038/s41589-022-01047-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 04/26/2022] [Indexed: 02/08/2023]
Abstract
Regenerating pancreatic β-cells is a potential curative approach for diabetes. We previously identified the small molecule CID661578 as a potent inducer of β-cell regeneration, but its target and mechanism of action have remained unknown. We now screened 257 million yeast clones and determined that CID661578 targets MAP kinase-interacting serine/threonine kinase 2 (MNK2), an interaction we genetically validated in vivo. CID661578 increased β-cell neogenesis from ductal cells in zebrafish, neonatal pig islet aggregates and human pancreatic ductal organoids. Mechanistically, we found that CID661578 boosts protein synthesis and regeneration by blocking MNK2 from binding eIF4G in the translation initiation complex at the mRNA cap. Unexpectedly, this blocking activity augmented eIF4E phosphorylation depending on MNK1 and bolstered the interaction between eIF4E and eIF4G, which is necessary for both hypertranslation and β-cell regeneration. Taken together, our findings demonstrate a targetable role of MNK2-controlled translation in β-cell regeneration, a role that warrants further investigation in diabetes.
Collapse
|
44
|
Weiss JM, Lumaquin-Yin D, Montal E, Suresh S, Leonhardt CS, White RM. Shifting the focus of zebrafish toward a model of the tumor microenvironment. eLife 2022; 11:69703. [PMID: 36538362 PMCID: PMC9767465 DOI: 10.7554/elife.69703] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/21/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer cells exist in a complex ecosystem with numerous other cell types in the tumor microenvironment (TME). The composition of this tumor/TME ecosystem will vary at each anatomic site and affects phenotypes such as initiation, metastasis, and drug resistance. A mechanistic understanding of the large number of cell-cell interactions between tumor and TME requires models that allow us to both characterize as well as genetically perturb this complexity. Zebrafish are a model system optimized for this problem, because of the large number of existing cell-type-specific drivers that can label nearly any cell in the TME. These include stromal cells, immune cells, and tissue resident normal cells. These cell-type-specific promoters/enhancers can be used to drive fluorophores to facilitate imaging and also CRISPR cassettes to facilitate perturbations. A major advantage of the zebrafish is the ease by which large numbers of TME cell types can be studied at once, within the same animal. While these features make the zebrafish well suited to investigate the TME, the model has important limitations, which we also discuss. In this review, we describe the existing toolset for studying the TME using zebrafish models of cancer and highlight unique biological insights that can be gained by leveraging this powerful resource.
Collapse
Affiliation(s)
- Joshua M Weiss
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Dianne Lumaquin-Yin
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Emily Montal
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Shruthy Suresh
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Carl S Leonhardt
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States,Department of Medicine, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
45
|
Schmitner N, Recheis C, Thönig J, Kimmel RA. Differential Responses of Neural Retina Progenitor Populations to Chronic Hyperglycemia. Cells 2021; 10:cells10113265. [PMID: 34831487 PMCID: PMC8622914 DOI: 10.3390/cells10113265] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/18/2021] [Indexed: 12/30/2022] Open
Abstract
Diabetic retinopathy is a frequent complication of longstanding diabetes, which comprises a complex interplay of microvascular abnormalities and neurodegeneration. Zebrafish harboring a homozygous mutation in the pancreatic transcription factor pdx1 display a diabetic phenotype with survival into adulthood, and are therefore uniquely suitable among zebrafish models for studying pathologies associated with persistent diabetic conditions. We have previously shown that, starting at three months of age, pdx1 mutants exhibit not only vascular but also neuro-retinal pathologies manifesting as photoreceptor dysfunction and loss, similar to human diabetic retinopathy. Here, we further characterize injury and regenerative responses and examine the effects on progenitor cell populations. Consistent with a negative impact of hyperglycemia on neurogenesis, stem cells of the ciliary marginal zone show an exacerbation of aging-related proliferative decline. In contrast to the robust Müller glial cell proliferation seen following acute retinal injury, the pdx1 mutant shows replenishment of both rod and cone photoreceptors from slow-cycling, neurod-expressing progenitors which first accumulate in the inner nuclear layer. Overall, we demonstrate a diabetic retinopathy model which shows pathological features of the human disease evolving alongside an ongoing restorative process that replaces lost photoreceptors, at the same time suggesting an unappreciated phenotypic continuum between multipotent and photoreceptor-committed progenitors.
Collapse
|
46
|
Markov DA, Petrucco L, Kist AM, Portugues R. A cerebellar internal model calibrates a feedback controller involved in sensorimotor control. Nat Commun 2021; 12:6694. [PMID: 34795244 PMCID: PMC8602262 DOI: 10.1038/s41467-021-26988-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/28/2021] [Indexed: 11/18/2022] Open
Abstract
Animals must adapt their behavior to survive in a changing environment. Behavioral adaptations can be evoked by two mechanisms: feedback control and internal-model-based control. Feedback controllers can maintain the sensory state of the animal at a desired level under different environmental conditions. In contrast, internal models learn the relationship between the motor output and its sensory consequences and can be used to recalibrate behaviors. Here, we present multiple unpredictable perturbations in visual feedback to larval zebrafish performing the optomotor response and show that they react to these perturbations through a feedback control mechanism. In contrast, if a perturbation is long-lasting, fish adapt their behavior by updating a cerebellum-dependent internal model. We use modelling and functional imaging to show that the neuronal requirements for these mechanisms are met in the larval zebrafish brain. Our results illustrate the role of the cerebellum in encoding internal models and how these can calibrate neuronal circuits involved in reactive behaviors depending on the interactions between animal and environment.
Collapse
Affiliation(s)
- Daniil A Markov
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany
| | - Luigi Petrucco
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany
- Institute of Neuroscience, Technical University of Munich, 80802, Munich, Germany
| | - Andreas M Kist
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany
- Division of Phoniatrics and Pediatric Audiology, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Ruben Portugues
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany.
- Institute of Neuroscience, Technical University of Munich, 80802, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
47
|
Rovira M, Atla G, Maestro MA, Grau V, García-Hurtado J, Maqueda M, Mosquera JL, Yamada Y, Kerr-Conte J, Pattou F, Ferrer J. REST is a major negative regulator of endocrine differentiation during pancreas organogenesis. Genes Dev 2021; 35:1229-1242. [PMID: 34385258 PMCID: PMC8415321 DOI: 10.1101/gad.348501.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022]
Abstract
In this study, Rovira et al. report that inactivation of the transcriptional repressor REST causes a drastic increase in pancreatic endocrine progenitors and endocrine cells, and establish that REST is a major negative regulator of embryonic pancreas endocrine differentiation in mice and zebrafish. Their findings show that REST-dependent inhibition ensures a balanced production of endocrine cells from embryonic pancreatic progenitors. Multiple transcription factors have been shown to promote pancreatic β-cell differentiation, yet much less is known about negative regulators. Earlier epigenomic studies suggested that the transcriptional repressor REST could be a suppressor of endocrinogenesis in the embryonic pancreas. However, pancreatic Rest knockout mice failed to show abnormal numbers of endocrine cells, suggesting that REST is not a major regulator of endocrine differentiation. Using a different conditional allele that enables profound REST inactivation, we observed a marked increase in pancreatic endocrine cell formation. REST inhibition also promoted endocrinogenesis in zebrafish and mouse early postnatal ducts and induced β-cell-specific genes in human adult duct-derived organoids. We also defined genomic sites that are bound and repressed by REST in the embryonic pancreas. Our findings show that REST-dependent inhibition ensures a balanced production of endocrine cells from embryonic pancreatic progenitors.
Collapse
Affiliation(s)
- Meritxell Rovira
- Department of Physiological Science, School of Medicine, Universitat de Barcelona (UB), L'Hospitalet de Llobregat, Barcelona 08907, Spain.,Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Barcelona 08908, Spain.,Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P-CMR[C]), L'Hospitalet de Llobregat, Barcelona 08908, Spain.,Center for Networked Biomedical Research on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Goutham Atla
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona 08003, Spain
| | - Miguel Angel Maestro
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona 08003, Spain.,Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid 28029, Spain
| | - Vane Grau
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona 08003, Spain.,Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid 28029, Spain
| | - Javier García-Hurtado
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona 08003, Spain.,Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid 28029, Spain
| | - Maria Maqueda
- Bioinformatics Unit, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Jose Luis Mosquera
- Bioinformatics Unit, Bellvitge Biomedical Research Institute, IDIBELL, L'Hospitalet del Llobregat, Barcelona 08908, Spain
| | - Yasuhiro Yamada
- Division of Stem Cell Pathology, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Julie Kerr-Conte
- Institute Pasteur Lille, University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire de Lille (CHU Lille), U1190, European Genomic Institute for Diabetes (EGID), Lille F-59000, France
| | - Francois Pattou
- Institute Pasteur Lille, University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire de Lille (CHU Lille), U1190, European Genomic Institute for Diabetes (EGID), Lille F-59000, France
| | - Jorge Ferrer
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona 08003, Spain.,Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid 28029, Spain.,Department of Metabolism, Digestion, and Reproduction, Section of Genetics and Genomics, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
48
|
Kulkarni A, Pineros AR, Walsh MA, Casimiro I, Ibrahim S, Hernandez-Perez M, Orr KS, Glenn L, Nadler JL, Morris MA, Tersey SA, Mirmira RG, Anderson RM. 12-Lipoxygenase governs the innate immune pathogenesis of islet inflammation and autoimmune diabetes. JCI Insight 2021; 6:e147812. [PMID: 34128835 PMCID: PMC8410073 DOI: 10.1172/jci.insight.147812] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Macrophages and related myeloid cells are innate immune cells that participate in the early islet inflammation of type 1 diabetes (T1D). The enzyme 12-lipoxygenase (12-LOX) catalyzes the formation of proinflammatory eicosanoids, but its role and mechanisms in myeloid cells in the pathogenesis of islet inflammation have not been elucidated. Leveraging a model of islet inflammation in zebrafish, we show here that macrophages contribute significantly to the loss of β cells and the subsequent development of hyperglycemia. The depletion or inhibition of 12-LOX in this model resulted in reduced macrophage infiltration into islets and the preservation of β cell mass. In NOD mice, the deletion of the gene encoding 12-LOX in the myeloid lineage resulted in reduced insulitis with reductions in proinflammatory macrophages, a suppressed T cell response, preserved β cell mass, and almost complete protection from the development of T1D. 12-LOX depletion caused a defect in myeloid cell migration, a function required for immune surveillance and tissue injury responses. This effect on migration resulted from the loss of the chemokine receptor CXCR3. Transgenic expression of the gene encoding CXCR3 rescued the migratory defect in zebrafish 12-LOX morphants. Taken together, our results reveal a formative role for innate immune cells in the early pathogenesis of T1D and identify 12-LOX as an enzyme required to promote their prodiabetogenic phenotype in the context of autoimmunity.
Collapse
Affiliation(s)
- Abhishek Kulkarni
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Annie R Pineros
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Melissa A Walsh
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Isabel Casimiro
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Sara Ibrahim
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marimar Hernandez-Perez
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kara S Orr
- Center for Diabetes and Metabolic Diseases and Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Lindsey Glenn
- Department of Medicine, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Jerry L Nadler
- Department of Medicine, New York Medical College, Valhalla, New York, USA
| | - Margaret A Morris
- Department of Medicine, Eastern Virginia Medical School, Norfolk, Virginia, USA
| | - Sarah A Tersey
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Raghavendra G Mirmira
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Ryan M Anderson
- Kolver Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
49
|
Kim E, Lee DW, Park HC, Kim DH. Protective effects of alpha-lipoic acid on hair cell damage in diabetic zebrafish model. Mol Genet Metab Rep 2021; 28:100783. [PMID: 34354927 PMCID: PMC8322127 DOI: 10.1016/j.ymgmr.2021.100783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 11/04/2022] Open
Abstract
Hearing impairment is one of the complications in diabetes mellitus; however, there are very few therapeutic studies on it. In this study, we investigated the protective effect of alpha-lipoic acid (ALA) on hearing loss in diabetic transgenic zebrafish and confirmed that ALA protects the loss of hair cells (HCs) caused by hyperglycemia. The data indicated that ALA has a protective effect on the damage to HCs in diabetic zebrafish.
Collapse
Affiliation(s)
- Eunmi Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Republic of Korea
| | - Dong-Won Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Republic of Korea
| | - Dong Hwee Kim
- Department of Physical Medicine and Rehabilitation, College of Medicine, Korea University, Ansan 15355, Republic of Korea
| |
Collapse
|
50
|
Characteristics of the New Insulin-Resistant Zebrafish Model. Pharmaceuticals (Basel) 2021; 14:ph14070642. [PMID: 34358068 PMCID: PMC8308799 DOI: 10.3390/ph14070642] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/09/2023] Open
Abstract
Insulin resistance, which occurs when insulin levels are sufficiently high over a prolonged period, causing the cells to fail to respond normally to the hormone. As a system for insulin resistance and diabetes drug development, insulin-resistant rodent models have been clearly established, but there is a limitation to high-throughput drug screening. Recently, zebrafish have been identified as an excellent system for drug discovery and identification of therapeutic targets, but studies on insulin resistance models have not been extensively performed. Therefore, we aimed to make a rapid insulin-resistant zebrafish model that complements the existing rodent models. To establish this model, zebrafish were treated with 10 μM insulin for 48 h. This model showed characteristics of insulin-resistant disease such as damaged pancreatic islets. Then we confirmed the recovery of the pancreatic islets after pioglitazone treatment. In addition, it was found that insulin-resistant drugs have as significant an effect in zebrafish as in humans, and these results proved the value of the zebrafish insulin resistance model for drug selection. In addition, RNA sequencing was performed to elucidate the mechanism involved. KEGG pathway enrichment analysis of differentially expressed genes showed that insulin resistance altered gene expression due to the MAPK signaling and calcium signaling pathways. This model demonstrates the utility of the zebrafish model for drug testing and drug discovery in insulin resistance and diabetes.
Collapse
|