1
|
Leon-Icaza SA, Frétaud M, Cornélie S, Bureau C, Yatime L, Floto RA, Renshaw SA, Herrmann JL, Langevin C, Cougoule C, Bernut A. Curcumin-mediated NRF2 induction limits inflammatory damage in, preclinical models of cystic fibrosis. Biomed Pharmacother 2025; 186:117957. [PMID: 40168724 DOI: 10.1016/j.biopha.2025.117957] [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: 12/21/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Overactive neutrophilic inflammation causes damage to the airways and death in people with cystic fibrosis (CF), a genetic disorder resulting from mutations in the CFTR gene. Reducing the impact of inflammation is therefore a major concern in CF. Evidence indicates that dysfunctional NRF2 signaling in CF individuals may impair their ability to regulate their oxidative and inflammatory responses, although the role of NRF2 in neutrophil-dominated inflammation and tissue damage associated with CF has not been determined. Therefore, we examined whether curcumin, an activator of NRF2, might provide a beneficial effect in the context of CF. METHODS Combining Cftr-depleted zebrafish as an innovative biomedical model with CF patient-derived airway organoids (AOs), we aimed to understand how NRF2 dysfunction leads to abnormal inflammatory status and tissue remodeling and determine the effects of curcumin in reducing inflammation and tissue damage in CF. RESULTS We demonstrate that NFR2 is instrumental in regulating neutrophilic inflammation and repair processes in vivo, thereby preventing inflammatory damage. Importantly, curcumin treatment restores NRF2 activity in both CF zebrafish and AOs. Curcumin reduces neutrophilic inflammation in CF context, by rebalancing the production of epithelial ROS and pro-inflammatory cytokines. Furthermore, curcumin improves tissue repair by reducing CF-associated fibrosis. Our findings demonstrate that curcumin prevents CF-mediated inflammation via activating the NRF2 pathway. CONCLUSIONS This work highlights the protective role of NRF2 in limiting inflammation and injury and show that therapeutic strategies to normalize NRF2 activity, using curcumin or others NRF2 activators, might simultaneously reduce airway inflammation and damage in CF.
Collapse
Affiliation(s)
- Stephen A Leon-Icaza
- Institute of Pharmacology and Structural Biology, University of Toulouse, CNRS, Toulouse, France
| | - Maxence Frétaud
- Université Paris-Saclay, INRAE, Université de Versailles St Quentin, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Sarahdja Cornélie
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Charlotte Bureau
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Laure Yatime
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - R Andres Floto
- Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC-Laboratory of Molecular Biology, Cambridge, UK
| | - Stephen A Renshaw
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Jean-Louis Herrmann
- Université Paris-Saclay, Université de Versailles St Quentin, Inserm, Infection et Inflammation, Montigny-le-Bretonneux, France
| | - Christelle Langevin
- Université Paris-Saclay, INRAE, Infectiologie Expérimentale des Rongeurs et des Poissons, Jouy-en-Josas, France
| | - Céline Cougoule
- Institute of Pharmacology and Structural Biology, University of Toulouse, CNRS, Toulouse, France
| | - Audrey Bernut
- Laboratory of Pathogens and Host Immunity, University of Montpellier, CNRS, Inserm, Montpellier, France.
| |
Collapse
|
2
|
Liang W, Hou C, Zhu Z, Wang P, Wang X, Li Z, Xue J, Ran R. Cutaneous Pigment Cell Distributions and Skin Structure of Xenopus. Pigment Cell Melanoma Res 2025; 38:e70022. [PMID: 40329555 DOI: 10.1111/pcmr.70022] [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: 02/06/2025] [Revised: 04/05/2025] [Accepted: 04/18/2025] [Indexed: 05/08/2025]
Abstract
Pigment cells not only are intrinsic factors to determine animal patterns, but also play vital roles in numerous behavioral and physiological processes as well as health, such as melanomas originating from melanocytes. Model organisms are commonly used to study pigment cell development and the mechanisms underlying related diseases, with zebrafish and mice, and Xenopus being well-established examples. Xenopus tropicalis, a diploid amphibian model, offers advantages such as high fecundity and easily observable pigment cell development. Recent advancements in gene-editing techniques have increased its prominence in research on pigment cell biology and melanoma pathogenesis. Here, we compare the skin pigment cell distribution as well as the skin structure in X. tropicalis, zebrafish, mice, and humans and point out the potential value of using X. tropicalis to model human skin diseases, such as melanoma.
Collapse
Affiliation(s)
- Weizheng Liang
- Hebei Key Laboratory of Systems Biology and Gene Regulation, Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | | | | | - Peng Wang
- Hebei North University, Zhangjiakou, China
| | - Xiran Wang
- Department of Bioinformatics, School of Health Care, Changchun Vocational College of Health, Changchun, China
| | - Zhongwu Li
- Department of Pathology, Peking University Cancer Hospital, Beijing, China
| | - Jun Xue
- Department of Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Rensen Ran
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University, Beijing, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
3
|
Li J, Liu MJ, Du WJ, Peng XL, Deng H, Zi HX, Shang HB, Du JL. Neural-activity-regulated and glia-mediated control of brain lymphatic development. Cell 2025:S0092-8674(25)00410-6. [PMID: 40311620 DOI: 10.1016/j.cell.2025.04.008] [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: 10/30/2024] [Revised: 01/28/2025] [Accepted: 04/03/2025] [Indexed: 05/03/2025]
Abstract
The nervous system regulates peripheral immune responses under physiological and pathological conditions, but the brain's impact on immune system development remains unknown. Meningeal mural lymphatic endothelial cells (muLECs), embedded in the leptomeninges, form an immune niche surrounding the brain that contributes to brain immunosurveillance. Here, we report that the brain controls the development of muLECs via a specialized glial subpopulation, slc6a11b+ radial astrocytes (RAs), a process modulated by neural activity in zebrafish. slc6a11b+ RAs, with processes extending to the meninges, govern muLEC formation by expressing vascular endothelial growth factor C (vegfc). Moreover, neural activity regulates muLEC development, and this regulation requires Vegfc in slc6a11b+ RAs. Intriguingly, slc6a11b+ RAs cooperate with calcium-binding EGF domain 1 (ccbe1)+ fibroblasts to restrict muLEC growth on the brain surface via controlling mature Vegfc distribution. Thus, our study uncovers a glia-mediated and neural-activity-regulated control of brain lymphatic development and highlights the importance of inter-tissue cellular cooperation in development.
Collapse
Affiliation(s)
- Jia Li
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Ming-Jian Liu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wen-Jie Du
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiao-Lan Peng
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hao Deng
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hua-Xing Zi
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Han-Bing Shang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Emergency Medicine Center, Shanghai Institute of Aviation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jiu-Lin Du
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-Inspired Intelligence Technology, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China.
| |
Collapse
|
4
|
Zheng L, Wu Z, Yada N, Liu S, Lin C, Bignotti A, Zhao X, Zheng XL. Modeling ANKRD26 5'-UTR mutation-related thrombocytopenia. Dis Model Mech 2025; 18:dmm052222. [PMID: 40170493 PMCID: PMC12067082 DOI: 10.1242/dmm.052222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/26/2025] [Indexed: 04/03/2025] Open
Abstract
Mutations in the 5'-untranslated region (5'-UTR) of ankyrin repeat domain-containing protein 26 (ANKRD26) are associated with hereditary thrombocytopenia 2 (THC2). However, the causative role of these mutations and the mechanisms underlying THC2 are not fully established. Here, we report, for the first time, that zebrafish carrying a deletion of two nucleotides (Δ2) in the 5'-UTR of ankrd26 recapitulate some of the key laboratory features of THC2. ankrd26ku6 (homozygous for the Δ2 deletion in the 5'-UTR) fish larvae exhibited significantly increased expression of ankrd26 mRNA and protein. Adult ankrd26ku6 fish exhibited spontaneous thrombocytopenia. Furthermore, the thrombocytes from ankrd26ku6 fish showed enhanced ability to adhere and aggregate on a collagen surface under flow. Proteomic profiling demonstrated marked upregulation of Ninjurin 1 in young thrombocytes from ankrd26ku6 fish compared with those from wild-type controls. The ankrd26ku6 fish with a homozygous nacre allele developed myelodysplastic syndrome at old age. ANKRD26 protein levels were also significantly increased in platelets and plasma from patients with immune thrombotic thrombocytopenic purpura compared with those from unaffected controls. We conclude that ANKRD26 overexpression, resulting from either hereditary or acquired mechanisms, contributes to thrombocytopenia, thrombosis and hematologic malignancies.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Zhijian Wu
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410010, China
| | - Noritaka Yada
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Szumam Liu
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Cindy Lin
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Antonia Bignotti
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Xinyang Zhao
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - X. Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| |
Collapse
|
5
|
Menard R, Baanannou A, Halluin C, Morse D, Kuhn S, Graber JH, Strickland J, Madelaine R. The small molecule ML233 is a direct inhibitor of tyrosinase function. Commun Biol 2025; 8:506. [PMID: 40155764 PMCID: PMC11953452 DOI: 10.1038/s42003-025-07973-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 03/20/2025] [Indexed: 04/01/2025] Open
Abstract
Melanogenesis is the biological process regulating the synthesis of melanin pigments in melanocytes. Defective melanogenesis is associated with numerous human skin diseases, including, but not limited to, albinism, vitiligo, melasma, and hypo- and hyperpigmentation disorders. Tyrosinase is the rate-limiting enzyme controlling melanogenesis, and hence tremendous efforts have been made to identify potent and safe inhibitors of tyrosinase function. However, despite decades of research, currently there is no effective treatment that inhibits melanogenesis or tyrosinase activity with no adverse side effects. In this study, we report the characterization of the ML233 chemical as a potent inhibitor of tyrosinase activity in vivo and in vitro. We demonstrate that ML233 reduces melanin production in the zebrafish model with no observable significant toxic side effects, and in murine melanoma cells. We also predict that these effects are mediated through direct tyrosinase-ML233 interaction, i.e., binding of the ML233 molecule to the active site of the protein to inhibit its function. Together, our results reveal that ML233 plays roles in both healthy and pathological skin cells via inhibition of melanin production. ML233-mediated tyrosinase inhibition is a potentially safe and effective approach to alleviate the symptoms of melanocyte-associated diseases and thereby substantially improve human health.
Collapse
Affiliation(s)
- Romain Menard
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, ME, USA
| | - Aissette Baanannou
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, ME, USA
| | - Caroline Halluin
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, ME, USA
| | - Dexter Morse
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, ME, USA
| | | | - Joel H Graber
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, ME, USA
| | | | - Romain Madelaine
- MDI Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Bar Harbor, ME, USA.
| |
Collapse
|
6
|
Montandon SA, Beaudier P, Ullate-Agote A, Helleboid PY, Kummrow M, Roig-Puiggros S, Jabaudon D, Andersson L, Milinkovitch MC, Tzika AC. Regulatory and disruptive variants in the CLCN2 gene are associated with modified skin color pattern phenotypes in the corn snake. Genome Biol 2025; 26:73. [PMID: 40140900 PMCID: PMC11948899 DOI: 10.1186/s13059-025-03539-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Snakes exhibit a broad variety of adaptive colors and color patterns, generated by the spatial arrangement of chromatophores, but little is known of the mechanisms responsible for these spectacular traits. Here, we investigate a mono-locus trait with two recessive alleles, motley and stripe, that both cause pattern aberrations in the corn snake. RESULTS We use mapping-by-sequencing to identify the genomic interval where the causal mutations reside. With our differential gene expression analyses, we find that CLCN2 (Chloride Voltage-Gated Channel 2), a gene within the genomic interval, is significantly downregulated in Motley embryonic skin. Furthermore, we identify the stripe allele as the insertion of an LTR-retrotransposon in CLCN2, resulting in a disruptive mutation of the protein. We confirm the involvement of CLCN2 in color pattern formation by producing knock-out snakes that present a phenotype similar to Stripe. In humans and mice, disruption of CLCN2 results in leukoencephalopathy, as well as retinal and testes degeneration. Our single-cell transcriptomic analyses in snakes reveal that CLCN2 is indeed expressed in chromatophores during embryogenesis and in the adult brain, but the behavior and fertility of Motley and Stripe corn snakes are not impacted. CONCLUSIONS Our genomic, transcriptomic, and functional analyses identify a plasma membrane anion channel to be involved in color pattern development in snakes and show that an active LTR-retrotransposon might be a key driver of trait diversification in corn snakes.
Collapse
Affiliation(s)
- Sophie A Montandon
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
- Present address: Bracco Suisse S.A., Plan-les-Ouates, Switzerland
| | - Pierre Beaudier
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
| | - Asier Ullate-Agote
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
- Present address: Biomedical Engineering Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Pierre-Yves Helleboid
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
| | - Maya Kummrow
- Tierspital, University of Zurich, Zurich, Switzerland
| | - Sergi Roig-Puiggros
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- Clinic of Neurology, Geneva University Hospital, Geneva, Switzerland
| | - Leif Andersson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Michel C Milinkovitch
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland.
| | - Athanasia C Tzika
- Laboratory of Artificial and Natural Evolution, Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
7
|
Zi H, Peng X, Du J, Li J. Protocol for generating a pericyte reporter zebrafish line Ki(pdgfrb-P2A-GAL4-VP16) using a CRISPR-Cas9-mediated knockin technique. STAR Protoc 2025; 6:103490. [PMID: 39673702 PMCID: PMC11699729 DOI: 10.1016/j.xpro.2024.103490] [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: 04/30/2024] [Revised: 08/29/2024] [Accepted: 11/08/2024] [Indexed: 12/16/2024] Open
Abstract
Pericytes, the mural cells that envelop small blood vessels, play crucial roles in the formation of the blood-brain barrier (BBB). Here, we present a protocol for generating a pericyte reporter zebrafish line Ki(pdgfrb-P2A-GAL4-VP16) using a CRISPR-Cas9-mediated knockin technique. We describe steps for identifying efficient single guide RNA (sgRNA), constructing donor plasmid, and generating and maintaining the knockin line. We then detail procedures for in vivo imaging of brain pericytes. This protocol is adaptable for creating other knockin lines for specific cell labeling. For complete details on the use and execution of this protocol, please refer to Zi et al.1.
Collapse
Affiliation(s)
- Huaxing Zi
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing 100049, China; Dongguan Innovation Institute, Guangdong Medical University, 1 Xin-Cheng Road, Dongguan, Guangdong 523808, China
| | - Xiaolan Peng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai 200031, China.
| | - Jia Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China.
| |
Collapse
|
8
|
Korzeniwsky KG, de Mello PL, Liang Y, Feltes M, Farber SA, Parichy DM. Dominant Negative Mitf Allele Impacts Melanophore and Xanthophore Development and Reveals Collaborative Interactions With Tfec in Zebrafish Chromatophore Lineages. Pigment Cell Melanoma Res 2025; 38:e70009. [PMID: 40123122 PMCID: PMC11931198 DOI: 10.1111/pcmr.70009] [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: 12/30/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/25/2025]
Abstract
Ectothermic vertebrates exhibit a diverse array of pigment cell types-chromatophores-that provide valuable opportunities to uncover mechanisms of fate specification and how they evolve. Like melanocytes of mammals, the melanophores of teleosts and other ectotherms depend on basic helix-loop-helix leucine zipper transcription factors encoded by orthologues of MITF. A different chromatophore, the iridescent iridophore, depends on the closely related transcription factor Tfec. Requirements for the specification of other chromatophore lineages remain largely uncertain. Here we identify a new allele of the zebrafish Mitf gene, mitfa, that results in a complete absence of not only melanophores but also yellow-orange xanthophores. Harboring a missense substitution in the DNA-binding domain identical to previously isolated alleles of mouse, we show that this new allele has defects in chromatophore precursor survival and xanthophore differentiation that extend beyond those of mitfa loss-of-function. Additional genetic analyses revealed interactions between Mitfa and Tfec as a likely basis for the observed phenotypes. Our findings point to collaborative roles for Mitfa and Tfec in promoting chromatophore development, particularly in xanthophore lineages, and provide new insights into evolutionary aspects of MITF functions across vertebrates.
Collapse
Affiliation(s)
| | | | - Yipeng Liang
- Department of BiologyUniversity of VirginiaVirginiaUSA
| | - McKenna Feltes
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Steven A. Farber
- Department of BiologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - David M. Parichy
- Department of BiologyUniversity of VirginiaVirginiaUSA
- Department of Cell BiologyUniversity of VirginiaVirginiaUSA
| |
Collapse
|
9
|
Kroll F, Donnelly J, Özcan GG, Mackay E, Rihel J. Behavioural pharmacology predicts disrupted signalling pathways and candidate therapeutics from zebrafish mutants of Alzheimer's disease risk genes. eLife 2025; 13:RP96839. [PMID: 39960847 PMCID: PMC11832171 DOI: 10.7554/elife.96839] [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] [Indexed: 02/20/2025] Open
Abstract
By exposing genes associated with disease, genomic studies provide hundreds of starting points that should lead to druggable processes. However, our ability to systematically translate these genomic findings into biological pathways remains limited. Here, we combine rapid loss-of-function mutagenesis of Alzheimer's risk genes and behavioural pharmacology in zebrafish to predict disrupted processes and candidate therapeutics. FramebyFrame, our expanded package for the analysis of larval behaviours, revealed that decreased night-time sleep was common to F0 knockouts of all four late-onset Alzheimer's risk genes tested. We developed an online tool, ZOLTAR, which compares any behavioural fingerprint to a library of fingerprints from larvae treated with 3677 compounds. ZOLTAR successfully predicted that sorl1 mutants have disrupted serotonin signalling and identified betamethasone as a drug which normalises the excessive day-time sleep of presenilin-2 knockout larvae with minimal side effects. Predictive behavioural pharmacology offers a general framework to rapidly link disease-associated genes to druggable pathways.
Collapse
Affiliation(s)
- François Kroll
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
- Institut de la Vision, Sorbonne UniversitéParisFrance
| | - Joshua Donnelly
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Güliz Gürel Özcan
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Eirinn Mackay
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| |
Collapse
|
10
|
Dowell CK, Hawkins T, Bianco IH. Subsets of extraocular motoneurons produce kinematically distinct saccades during hunting and exploration. Curr Biol 2025; 35:554-573.e6. [PMID: 39818217 DOI: 10.1016/j.cub.2024.12.010] [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: 09/20/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/18/2025]
Abstract
Animals construct diverse behavioral repertoires by moving a limited number of body parts with varied kinematics and patterns of coordination. There is evidence that distinct movements can be generated by changes in activity dynamics within a common pool of motoneurons or by selectively engaging specific subsets of motoneurons in a task-dependent manner. However, in most cases, we have an incomplete understanding of the patterns of motoneuron activity that generate distinct actions and of how upstream premotor circuits select and assemble such motor programs. In this study, we used two closely related but kinematically distinct types of saccadic eye movement in larval zebrafish as a model to examine circuit control of movement diversity. In contrast to the prevailing view of a final common pathway, we found that in the oculomotor nucleus, distinct subsets of motoneurons were engaged for each saccade type. This type-specific recruitment was topographically organized and aligned with ultrastructural differences in motoneuron morphology and afferent synaptic innervation. Medially located motoneurons were active for both saccade types, and circuit tracing revealed a type-agnostic premotor pathway that appears to control their recruitment. By contrast, a laterally located subset of motoneurons was specifically active for hunting-associated saccades and received premotor input from pretectal hunting command neurons. Our data support a model in which generalist and action-specific premotor pathways engage distinct subsets of motoneurons to elicit varied movements of the same body part that subserve distinct behavioral functions.
Collapse
Affiliation(s)
- Charles K Dowell
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - Thomas Hawkins
- Department of Cell & Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Isaac H Bianco
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
11
|
Zhang Y, Bai L, Wang X, Zhao Y, Zhang T, Ye L, Du X, Zhang Z, Du J, Wang K. Super-resolution imaging of fast morphological dynamics of neurons in behaving animals. Nat Methods 2025; 22:177-186. [PMID: 39578627 DOI: 10.1038/s41592-024-02535-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/24/2024] [Indexed: 11/24/2024]
Abstract
Neurons are best studied in their native states in which their functional and morphological dynamics support animals' natural behaviors. Super-resolution microscopy can potentially reveal these dynamics in higher details but has been challenging in behaving animals due to severe motion artifacts. Here we report multiplexed, line-scanning, structured illumination microscopy, which can tolerate motion of up to 50 μm s-1 while achieving 150-nm and 100-nm lateral resolutions in its linear and nonlinear forms, respectively. We continuously imaged the dynamics of spinules in dendritic spines and axonal boutons volumetrically over thousands of frames and tens of minutes in head-fixed mouse brains during sleep-wake cycles. Super-resolution imaging of axonal boutons revealed spinule dynamics on a scale of seconds. Simultaneous two-color imaging further enabled analyses of the spatial distributions of diverse PSD-95 clusters and opened up possibilities to study their correlations with the structural dynamics of dendrites in the brains of head-fixed awake mice.
Collapse
Affiliation(s)
- Yujie Zhang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lu Bai
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Wang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuchen Zhao
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tianlei Zhang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lichen Ye
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xufei Du
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhe Zhang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiulin Du
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kai Wang
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
12
|
Chen Y, Li H, Wang J, Yang S, Su Z, Wang W, Rao C, Hou L. The Ednrb-Aim2-AKT axis regulates neural crest-derived melanoblast proliferation during early development. Development 2024; 151:dev202444. [PMID: 39555938 DOI: 10.1242/dev.202444] [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: 10/26/2023] [Accepted: 10/17/2024] [Indexed: 11/19/2024]
Abstract
Ednrb is specifically required to develop neural crest (NC) stem cell-derived lineages. However, it is still unknown why Ednrb signaling is only needed for the early development of melanoblasts in the skin and eye. We show that Ednrb is required for the proliferation of melanoblasts during early mouse development. To understand the mechanism of melanoblast proliferation, we found that the gene absent in melanoma 2 (Aim2) is upregulated in Ednrb-deficient NC cells by RNA-sequencing analysis. Consequently, the knockdown or knockout of Aim2 partially rescued the proliferation of Ednrb-deficient melanoblasts. Conversely, the overexpression of Aim2 in melanoblasts suppressed their proliferation. We further show that Ednrb signaling could act through the microRNA miR-196b to block the suppression of melanoblast proliferation by Aim2 in primary NC cell cultures. These results reveal the Ednrb-Aim2-AKT axis in regulating melanocyte development and suggest that Ednrb signaling functions as a negative regulator of Aim2, which inhibits the proliferation of melanoblasts in early development. These findings uncover a previously unreported role for Aim2 outside the inflammasome, showing that it is a significant regulator controlling NC stem cell-derived lineage development.
Collapse
Affiliation(s)
- Yu Chen
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Huirong Li
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jing Wang
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shanshan Yang
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhongyuan Su
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wanxiao Wang
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Chunbao Rao
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
13
|
Sy SKH, Ko H. Fish-on-Chips: unveiling neural processing of chemicals in small animals through precise fluidic control. Neural Regen Res 2024; 19:2351-2353. [PMID: 38526270 PMCID: PMC11090447 DOI: 10.4103/1673-5374.392876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/05/2023] [Indexed: 03/26/2024] Open
Affiliation(s)
- Samuel K H Sy
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China (Sy SKH, Ko H)
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China (Sy SKH, Ko H)
- Department of Electrical and Electronic Engineering, Faculty of Engineering, The University of Hong Kong, Pok Fu Lam, Hong Kong Island, Hong Kong Special Administrative Region, China (Sy SKH)
- Advanced Biomedical Instrumentation Center, Hong Kong Science Park, Pak Shek Kok, New Territories, Hong Kong Special Administrative Region, China (Sy SKH)
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China (Sy SKH, Ko H)
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China (Sy SKH, Ko H)
- Margaret K. L. Cheung Research Center for Management of Parkinsonism, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China (Ko H)
- Lau Tat-chuen Research Center of Brain Degenerative Diseases in Chinese, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China (Ko H)
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region, China (Ko H)
| |
Collapse
|
14
|
McArthur KL, Ho WJ. Structure and Topography of Facial Branchiomotor Neuron Dendrites in Larval Zebrafish (Danio rerio). J Comp Neurol 2024; 532:e25682. [PMID: 39497493 PMCID: PMC11575941 DOI: 10.1002/cne.25682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/16/2024] [Indexed: 11/21/2024]
Abstract
Motor circuits in the vertebrate hindbrain need to become functional early in development. What are the fundamental mechanisms that establish early synaptic inputs to motor neurons? Previous evidence is consistent with the hypothesis that motor neuron dendrite positioning serves a causal role in early spinal motor circuit development, with initial connectivity determined by the overlap between premotor axons and motor neuron dendrites (perhaps without the need for molecular recognition). Does motor neuron dendrite topography serve a similar role in the hindbrain? In the current study, we provide the first quantitative analysis of the dendrites of facial branchiomotor neurons (FBMNs) in larval zebrafish. We previously demonstrated that FBMNs exhibit functional topography along the dorsoventral axis, with the most ventral cell bodies most likely to exhibit early rhythmic activity-suggesting that FBMNs with ventral cell bodies are most likely to receive inputs from premotor neurons carrying rhythmic respiratory signals. We hypothesized that this functional topography can be explained by differences in dendrite positioning, giving ventral FBMNs preferential access to premotor axons carrying rhythmic signals. If this hypothesis is true, we predicted that FBMN cell body position would be correlated with dendrite position along the dorsoventral axis. To test this prediction, we used single-cell labeling to trace the dendritic arbors of FBMNs in larval zebrafish at 5-days post-fertilization (dpf). FBMN dendrites varied in complexity, and this variation could not be attributed to differences in the relative age of neurons. Most dendrites grew caudally, laterally, and ventrally from the cell body-though FBMN dendrites could extend their dendrites dorsally. Across our sample, FBMN cell body position correlated with dendrite position along the dorsoventral axis, consistent with our hypothesis that differences in dendrite positioning serve as the substrate for differences in activity patterns across neurons. Future studies will build on this foundational data, testing additional predictions of the central hypothesis-to further investigate the mechanisms of early motor circuit development.
Collapse
Affiliation(s)
| | - Winnie J Ho
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| |
Collapse
|
15
|
Dowell CK, Lau JYN, Antinucci P, Bianco IH. Kinematically distinct saccades are used in a context-dependent manner by larval zebrafish. Curr Biol 2024; 34:4382-4396.e5. [PMID: 39236716 DOI: 10.1016/j.cub.2024.08.008] [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: 11/07/2023] [Revised: 06/27/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024]
Abstract
Saccades are rapid eye movements that are used by all species with good vision. In this study, we set out to characterize the complete repertoire of larval zebrafish horizontal saccades to gain insight into their contributions to visually guided behavior and underlying neural control. We identified five saccade types, defined by systematic differences in kinematics and binocular coordination, which were differentially expressed across a variety of behavioral contexts. Conjugate saccades formed a large group that serves at least four functions. These include fast phases of the optokinetic nystagmus, visual scanning in stationary animals, and shifting gaze in coordination with body turns. In addition, we discovered a previously undescribed pattern of eye-body coordination in which small conjugate saccades partially oppose head rotation to maintain gaze during forward locomotion. Convergent saccades were coordinated with body movements to foveate prey targets during hunting. Detailed kinematic analysis showed that conjugate and convergent saccades differed in the millisecond coordination of the eyes and body and followed distinct velocity main sequence relationships. This challenges the prevailing view that all horizontal saccades are controlled by a common brainstem circuit and instead indicates saccade-type-specific neural control.
Collapse
Affiliation(s)
- Charles K Dowell
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - Joanna Y N Lau
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - Paride Antinucci
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK
| | - Isaac H Bianco
- Department of Neuroscience, Physiology & Pharmacology, UCL, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
16
|
Tayran H, Yilmaz E, Bhattarai P, Min Y, Wang X, Ma Y, Wang N, Jeong I, Nelson N, Kassara N, Cosacak MI, Dogru RM, Reyes-Dumeyer D, Stenersen JM, Reddy JS, Qiao M, Flaherty D, Gunasekaran TI, Yang Z, Jurisch-Yaksi N, Teich AF, Kanekiyo T, Tosto G, Vardarajan BN, İş Ö, Ertekin-Taner N, Mayeux R, Kizil C. ABCA7-dependent induction of neuropeptide Y is required for synaptic resilience in Alzheimer's disease through BDNF/NGFR signaling. CELL GENOMICS 2024; 4:100642. [PMID: 39216475 PMCID: PMC11480862 DOI: 10.1016/j.xgen.2024.100642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 05/04/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Genetic variants in ABCA7, an Alzheimer's disease (AD)-associated gene, elevate AD risk, yet its functional relevance to the etiology is unclear. We generated a CRISPR-Cas9-mediated abca7 knockout zebrafish to explore ABCA7's role in AD. Single-cell transcriptomics in heterozygous abca7+/- knockout combined with Aβ42 toxicity revealed that ABCA7 is crucial for neuropeptide Y (NPY), brain-derived neurotrophic factor (BDNF), and nerve growth factor receptor (NGFR) expressions, which are crucial for synaptic integrity, astroglial proliferation, and microglial prevalence. Impaired NPY induction decreased BDNF and synaptic density, which are rescuable with ectopic NPY. In induced pluripotent stem cell-derived human neurons exposed to Aβ42, ABCA7-/- suppresses NPY. Clinical data showed reduced NPY in AD correlated with elevated Braak stages, genetic variants in NPY associated with AD, and epigenetic changes in NPY, NGFR, and BDNF promoters linked to ABCA7 variants. Therefore, ABCA7-dependent NPY signaling via BDNF-NGFR maintains synaptic integrity, implicating its impairment in increased AD risk through reduced brain resilience.
Collapse
Affiliation(s)
- Hüseyin Tayran
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Elanur Yilmaz
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Prabesh Bhattarai
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Yuhao Min
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Xue Wang
- Department of Quantitative Health Sciences, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Yiyi Ma
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Ni Wang
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Inyoung Jeong
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nastasia Nelson
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Nada Kassara
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE), Tatzberg 41, 01307 Dresden, Germany
| | - Ruya Merve Dogru
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Dolly Reyes-Dumeyer
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Jakob Mørkved Stenersen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Joseph S Reddy
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Min Qiao
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Delaney Flaherty
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Tamil Iniyan Gunasekaran
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Zikun Yang
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andrew F Teich
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA; Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Giuseppe Tosto
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Badri N Vardarajan
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Özkan İş
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL 32224, USA; Department of Neurology, Mayo Clinic Florida, Jacksonville, FL 32224, USA
| | - Richard Mayeux
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA; Department of Psychiatry, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University Irving Medical Center, Columbia University, 722 W. 168th St., New York, NY 10032, USA
| | - Caghan Kizil
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; The Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
17
|
Tzika AC, Ullate-Agote A, Helleboid PY, Kummrow M. PMEL is involved in snake colour pattern transition from blotches to stripes. Nat Commun 2024; 15:7655. [PMID: 39227572 PMCID: PMC11371805 DOI: 10.1038/s41467-024-51927-0] [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: 04/15/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
Corn snakes are emerging models for animal colouration studies. Here, we focus on the Terrazzo morph, whose skin pattern is characterized by stripes rather than blotches. Using genome mapping, we discover a disruptive mutation in the coding region of the Premelanosome protein (PMEL) gene. Our transcriptomic analyses reveal that PMEL expression is significantly downregulated in Terrazzo embryonic tissues. We produce corn snake PMEL knockouts, which present a comparable colouration phenotype to Terrazzo and the subcellular structure of their melanosomes and xanthosomes is also similarly impacted. Our single-cell expression analyses of wild-type embryonic dorsal skin demonstrate that all chromatophore progenitors express PMEL at varying levels. Finally, we show that in wild-type embryos PMEL-expressing cells are initially uniformly spread before forming aggregates and eventually blotches, as seen in the adults. In Terrazzo embryos, the aggregates fail to form. Our results provide insights into the mechanisms governing colouration patterning in reptiles.
Collapse
Affiliation(s)
- Athanasia C Tzika
- Laboratory of Artificial & Natural Evolution (LANE), Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland.
| | - Asier Ullate-Agote
- Laboratory of Artificial & Natural Evolution (LANE), Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
- Biomedical Engineering Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Pierre-Yves Helleboid
- Laboratory of Artificial & Natural Evolution (LANE), Department of Genetics & Evolution, University of Geneva, Geneva, Switzerland
| | - Maya Kummrow
- Tierspital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Kelsh RN. Myron Gordon Award Lecture 2023: Painting the neural crest: How studying pigment cells illuminates neural crest cell biology. Pigment Cell Melanoma Res 2024; 37:555-561. [PMID: 38010612 DOI: 10.1111/pcmr.13147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/28/2023] [Indexed: 11/29/2023]
Abstract
It has been 30 (!!) years since I began working on zebrafish pigment cells, as a postdoc in the laboratory of Prof. Christiane Nüsslein-Volhard. There, I participated in the first large-scale mutagenesis screen in zebrafish, focusing on pigment cell mutant phenotypes. The isolation of colourless, shady, parade and choker mutants allowed us (as a postdoc in Prof. Judith Eisen's laboratory, and then in my own laboratory at the University of Bath since 1997) to pursue my ambition to address long-standing problems in the neural crest field. Thus, we have studied how neural crest cells choose individual fates, resulting in our recent proposal of a new, and potentially unifying, model which we call Cyclical Fate Restriction, as well as addressing how pigment cell patterns are generated. A key feature of our work in the last 10 years has been the use of mathematical modelling approaches to clarify our biological models and to refine our interpretations. None of this would have been possible without a hugely talented group of laboratory members and other collaborators from around the world-it has been, and I am sure will continue to be, a pleasure and privilege to work with you all!
Collapse
Affiliation(s)
- Robert N Kelsh
- Department of Life Sciences, University of Bath, Bath, UK
| |
Collapse
|
19
|
Robinson CD, Milnes MR, Clifton IT, John-Alder HB, Cox RM. Evolutionary Loss of Male-Specific Coloration Is Associated with the Loss of Androgen Receptor Expression in Skin of Sceloporus Lizards. ECOLOGICAL AND EVOLUTIONARY PHYSIOLOGY 2024; 97:315-325. [PMID: 39680903 DOI: 10.1086/732782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
AbstractHormones can induce trait development in one species yet have no effect on the same trait in a closely related species, but the mechanisms underlying these differences are unclear. Here, we compare two closely related lizard species to explore the cellular mechanisms associated with the evolutionary loss of hormonally mediated ventral coloration. The eastern fence lizard (Sceloporus undulatus) has sexually dimorphic blue and black ventral coloration that develops when maturational increases in androgens induce melanin synthesis in males. The closely related striped plateau lizard (Sceloporus virgatus) has sexually monomorphic white ventral skin that does not produce melanin in response to the same signal. We used immunohistochemistry to localize the androgen receptor (AR) in the skin of both species and to test whether the loss of ventral coloration in S. virgatus corresponds to the loss of AR in the skin. We found that the ventral skin of S. virgatus displays little or no AR staining in the pigment cell layer, potentially explaining the loss of androgen sensitivity in this tissue, relative to the robust AR staining in the same layer of S. undulatus. Based on the location of three markers for melanophores (microphthalmia-associated transcription factor, dopachrome tautomerase, and tyrosinase), AR appears to be present in melanophores in S. undulatus. However, we could not detect these melanophore markers in the skin of S. virgatus. Therefore, the evolutionary loss of ventral coloration may have occurred via the loss of the AR-producing melanophore in mature ventral skin, preventing the development of a male-typical trait and sexual dimorphism in this tissue.
Collapse
|
20
|
Beaulieu MO, Thomas ED, Raible DW. Transdifferentiation is temporally uncoupled from progenitor pool expansion during hair cell regeneration in the zebrafish inner ear. Development 2024; 151:dev202944. [PMID: 39045613 PMCID: PMC11361639 DOI: 10.1242/dev.202944] [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: 04/09/2024] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
Death of mechanosensory hair cells in the inner ear is a common cause of auditory and vestibular impairment in mammals, which have a limited ability to regrow these cells after damage. In contrast, non-mammalian vertebrates, including zebrafish, can robustly regenerate hair cells after severe organ damage. The zebrafish inner ear provides an understudied model system for understanding hair cell regeneration in organs that are highly conserved with their mammalian counterparts. Here, we quantitatively examine hair cell addition during growth and regeneration of the larval zebrafish inner ear. We used a genetically encoded ablation method to induce hair cell death and we observed gradual regeneration with correct spatial patterning over a 2-week period following ablation. Supporting cells, which surround and are a source of new hair cells, divide in response to hair cell ablation, expanding the possible progenitor pool. In parallel, nascent hair cells arise from direct transdifferentiation of progenitor pool cells temporally uncoupled from supporting cell division. These findings reveal a previously unrecognized mechanism of hair cell regeneration with implications for how hair cells may be encouraged to regenerate in the mammalian ear.
Collapse
Affiliation(s)
- Marielle O. Beaulieu
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
| | - Eric D. Thomas
- Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - David W. Raible
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology - Head and Neck Surgery, University of Washington, Seattle, WA 98195, USA
- Neuroscience Graduate Program, University of Washington, Seattle, WA 98195, USA
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
21
|
Souto S, Lama R, Mérour E, Mehraz M, Bernard J, Lamoureux A, Massaad S, Frétaud M, Rigaudeau D, Millet JK, Langevin C, Biacchesi S. In vivo multiscale analyses of spring viremia of carp virus (SVCV) infection: From model organism to target species. PLoS Pathog 2024; 20:e1012328. [PMID: 39102417 PMCID: PMC11326706 DOI: 10.1371/journal.ppat.1012328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 08/15/2024] [Accepted: 06/07/2024] [Indexed: 08/07/2024] Open
Abstract
Spring viremia of carp virus (SVCV) has a broad fish host spectrum and is responsible for a disease that generally affects juvenile fishes with a mortality rate of up to 90%. In the absence of treatments or vaccines against SVCV, the search for prophylactic or therapeutic solutions is thus relevant, particularly to identify solutions compatible with mass vaccination. In addition to being a threat to aquaculture and ecosystems, SVCV is a unique pathogen to study virus-host interactions in the zebrafish model. Establishing the first reverse genetics system for SVCV and the design of recombinant SVCV (rSVCV) expressing fluorescent or bioluminescent proteins adds a new dimension for the study of these interactions using innovative imaging techniques. The infection by bath immersion of zebrafish larvae with rSVCV expressing mCherry allows us to define the first SVCV replication sites and the host innate immune responses using different transgenic lines of zebrafish. The fins were found as the main initial sites of infection in both zebrafish and carp, its natural host. Hence, new insights into the physiopathology of SVCV infection have been described. We report that neutrophils are recruited at the sites of infection and persist up to the death of the animal leading to an uncontrolled inflammation correlated with the expression of the pro-inflammatory cytokine IL1β. Tissue damage was observed at the site of initial replication, a likely consequence of virus-induced injury or the pro-inflammatory response. Interestingly, SVCV infection by bath immersion triggers a persistent pro-inflammatory response rather than activation of the antiviral IFN signaling pathway as observed following intravenous injection, highlighting the importance of the route of infection on the progression of pathogenicity. Thus, this model of zebrafish larvae infection by rSVCV offers new perspectives to study in detail virus-host interactions and to discover new prophylactic or therapeutic solutions.
Collapse
Affiliation(s)
- Sandra Souto
- Microbiology and Parasitology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Raquel Lama
- Microbiology and Parasitology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Emilie Mérour
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Manon Mehraz
- Université Paris-Saclay, INRAE, Infectiologie Expérimentale des Rongeurs et des Poissons, Jouy-en-Josas, France
| | - Julie Bernard
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Annie Lamoureux
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Sarah Massaad
- Université Paris-Saclay, INRAE, Infectiologie Expérimentale des Rongeurs et des Poissons, Jouy-en-Josas, France
| | - Maxence Frétaud
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Dimitri Rigaudeau
- Université Paris-Saclay, INRAE, Infectiologie Expérimentale des Rongeurs et des Poissons, Jouy-en-Josas, France
| | - Jean K Millet
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| | - Christelle Langevin
- Université Paris-Saclay, INRAE, Infectiologie Expérimentale des Rongeurs et des Poissons, Jouy-en-Josas, France
| | - Stéphane Biacchesi
- Université Paris-Saclay, INRAE, UVSQ, Virologie et Immunologie Moléculaires, Jouy-en-Josas, France
| |
Collapse
|
22
|
Xu Y, Han Y, Liu L, Han S, Zou S, Cheng B, Wang F, Xie X, Liang Y, Song M, Pang S. Highly sensitive response to the toxicity of environmental chemicals in transparent casper zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 948:174865. [PMID: 39032757 DOI: 10.1016/j.scitotenv.2024.174865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/15/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
The response sensitivity to toxic substances is the most concerned performance of animal model in chemical risk assessment. Casper (mitfaw2/w2;mpv17a9/a9), a transparent zebrafish mutant, is a useful in vivo model for toxicological assessment. However, the ability of casper to respond to the toxicity of exogenous chemicals is unknown. In this study, zebrafish embryos were exposed to five environmental chemicals, chlorpyrifos, lindane, α-endosulfan, bisphenol A, tetrabromobisphenol A (TBBPA), and an antiepileptic drug valproic acid. The half-lethal concentration (LC50) values of these chemicals in casper embryos were 62-87 % of that in the wild-type. After TBBPA exposure, the occurrence of developmental defects in the posterior blood island of casper embryos was increased by 67-77 % in relative to the wild-type, and the half-maximal effective concentration (EC50) in casper was 73 % of that in the wild-type. Moreover, the casper genetic background significantly increased the hyperlocomotion caused by chlorpyrifos and lindane exposure compared with the wild-type. These results demonstrated that casper had greater susceptibility to toxicity than wild-type zebrafish in acute toxicity, developmental toxicity and neurobehavioral toxicity assessments. Our data will inform future toxicological studies in casper and accelerate the development of efficient approaches and strategies for toxicity assessment via the use of casper.
Collapse
Affiliation(s)
- Yingjun Xu
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China; School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yiming Han
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Li Liu
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China; School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Shanshan Han
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Shibiao Zou
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Bo Cheng
- School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Fengbang Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Xunwei Xie
- China Zebrafish Resource Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Yong Liang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Maoyong Song
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
| | - Shaochen Pang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan 430056, China; School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, Wuhan 430205, China.
| |
Collapse
|
23
|
Elleman AV, Milicic N, Williams DJ, Simko J, Liu CJ, Haynes AL, Ehrlich DE, Makinson CD, Du Bois J. Behavioral control through the direct, focal silencing of neuronal activity. Cell Chem Biol 2024; 31:1324-1335.e20. [PMID: 38729162 PMCID: PMC11260259 DOI: 10.1016/j.chembiol.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/02/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024]
Abstract
The ability to optically stimulate and inhibit neurons has revolutionized neuroscience research. Here, we present a direct, potent, user-friendly chemical approach for optically silencing neurons. We have rendered saxitoxin (STX), a naturally occurring paralytic agent, transiently inert through chemical protection with a previously undisclosed nitrobenzyl-derived photocleavable group. Exposing the caged toxin, STX-bpc, to a brief (5 ms) pulse of light effects rapid release of a potent STX derivative and transient, spatially precise blockade of voltage-gated sodium channels (NaVs). We demonstrate the efficacy of STX-bpc for parametrically manipulating action potentials in mammalian neurons and brain slice. Additionally, we show the effectiveness of this reagent for silencing neural activity by dissecting sensory-evoked swimming in larval zebrafish. Photo-uncaging of STX-bpc is a straightforward method for non-invasive, reversible, spatiotemporally precise neural silencing without the need for genetic access, thus removing barriers for comparative research.
Collapse
Affiliation(s)
- Anna V Elleman
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA
| | - Nikola Milicic
- Department of Integrative Biology, University of Wisconsin-Madison, 121 Integrative Biology Research Building, 1117 W Johnson St, Madison, WI 53706, USA
| | - Damian J Williams
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA
| | - Jane Simko
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA
| | - Christine J Liu
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA; Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, MC 9872, New York, NY 10027, USA
| | - Allison L Haynes
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA
| | - David E Ehrlich
- Department of Integrative Biology, University of Wisconsin-Madison, 121 Integrative Biology Research Building, 1117 W Johnson St, Madison, WI 53706, USA.
| | - Christopher D Makinson
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 710 W 168th St, New York, NY 10032, USA; Department of Neuroscience, Columbia University, Jerome L. Greene Science Center, 3227 Broadway, MC 9872, New York, NY 10027, USA.
| | - J Du Bois
- Department of Chemistry, Stanford University, 333 Campus Drive, Stanford, CA 94305, USA.
| |
Collapse
|
24
|
Jeong I, Andreassen SN, Hoang L, Poulain M, Seo Y, Park HC, Fürthauer M, MacAulay N, Jurisch-Yaksi N. The evolutionarily conserved choroid plexus contributes to the homeostasis of brain ventricles in zebrafish. Cell Rep 2024; 43:114331. [PMID: 38843394 DOI: 10.1016/j.celrep.2024.114331] [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: 11/14/2023] [Revised: 04/24/2024] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
The choroid plexus (ChP) produces cerebrospinal fluid (CSF). It also contributes to brain development and serves as the CSF-blood barrier. Prior studies have identified transporters on the epithelial cells that transport water and ions from the blood vasculature to the ventricles and tight junctions involved in the CSF-blood barrier. Yet, how the ChP epithelial cells control brain physiology remains unresolved. We use zebrafish to provide insights into the physiological roles of the ChP. Upon histological and transcriptomic analyses, we identify that the zebrafish ChP is conserved with mammals and expresses transporters involved in CSF secretion. Next, we show that the ChP epithelial cells secrete proteins into CSF. By ablating the ChP epithelial cells, we identify a reduction of the ventricular sizes without alterations of the CSF-blood barrier. Altogether, our findings reveal that the zebrafish ChP is conserved and contributes to the size and homeostasis of the brain ventricles.
Collapse
Affiliation(s)
- Inyoung Jeong
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgsons Gate 1, 7491 Trondheim, Norway
| | - Søren N Andreassen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Linh Hoang
- Cellular and Molecular Imaging Core Facility (CMIC), Norwegian University of Science and Technology, Erling Skjalgsons Gate 1, 7491 Trondheim, Norway
| | - Morgane Poulain
- Université Côte d'Azur, CNRS, Inserm, iBV, 28 Avenue Valrose, 06108 Nice cedex 2, France
| | - Yongbo Seo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Maximilian Fürthauer
- Université Côte d'Azur, CNRS, Inserm, iBV, 28 Avenue Valrose, 06108 Nice cedex 2, France
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skjalgsons Gate 1, 7491 Trondheim, Norway.
| |
Collapse
|
25
|
Noonan HR, Thornock AM, Barbano J, Xifaras ME, Baron CS, Yang S, Koczirka K, McConnell AM, Zon LI. A chronic signaling TGFb zebrafish reporter identifies immune response in melanoma. eLife 2024; 13:e83527. [PMID: 38874379 PMCID: PMC11178360 DOI: 10.7554/elife.83527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 04/15/2024] [Indexed: 06/15/2024] Open
Abstract
Developmental signaling pathways associated with growth factors such as TGFb are commonly dysregulated in melanoma. Here we identified a human TGFb enhancer specifically activated in melanoma cells treated with TGFB1 ligand. We generated stable transgenic zebrafish with this TGFb Induced Enhancer driving green fluorescent protein (TIE:EGFP). TIE:EGFP was not expressed in normal melanocytes or early melanomas but was expressed in spatially distinct regions of advanced melanomas. Single-cell RNA-sequencing revealed that TIE:EGFP+ melanoma cells down-regulated interferon response while up-regulating a novel set of chronic TGFb target genes. ChIP-sequencing demonstrated that AP-1 factor binding is required for activation of chronic TGFb response. Overexpression of SATB2, a chromatin remodeler associated with tumor spreading, showed activation of TGFb signaling in early melanomas. Confocal imaging and flow cytometric analysis showed that macrophages localize to TIE:EGFP+ regions and preferentially phagocytose TIE:EGFP+ melanoma cells compared to TIE:EGFP- melanoma cells. This work identifies a TGFb induced immune response and demonstrates the need for the development of chronic TGFb biomarkers to predict patient response to TGFb inhibitors.
Collapse
Affiliation(s)
- Haley R Noonan
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
- Stem Cell and Regenerative Biology Department, Harvard UniversityCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
- Biological and Biomedical Sciences Program, Harvard Medical SchoolBostonUnited States
| | - Alexandra M Thornock
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
- Stem Cell and Regenerative Biology Department, Harvard UniversityCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
- Biological and Biomedical Sciences Program, Harvard Medical SchoolBostonUnited States
| | - Julia Barbano
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
| | - Michael E Xifaras
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
- Stem Cell and Regenerative Biology Department, Harvard UniversityCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
- Immunology Program, Harvard Medical SchoolBostonUnited States
| | - Chloe S Baron
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
- Stem Cell and Regenerative Biology Department, Harvard UniversityCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
| | - Song Yang
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
- Stem Cell and Regenerative Biology Department, Harvard UniversityCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
| | - Katherine Koczirka
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
| | - Alicia M McConnell
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
- Stem Cell and Regenerative Biology Department, Harvard UniversityCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children’s Hospital and Dana Farber Cancer Institute, Howard Hughes Medical InstituteBostonUnited States
- Stem Cell and Regenerative Biology Department, Harvard UniversityCambridgeUnited States
- Harvard Medical SchoolBostonUnited States
| |
Collapse
|
26
|
Tang S, Janpoom S, Prasertlux S, Rongmung P, Ittarat W, Ratdee O, Khamnamtong B, Klinbunga S. Identification of pigmentation genes in skin, muscle and tail of a Thai-flag variety of Siamese fighting fish Betta splendens. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 50:101243. [PMID: 38749208 DOI: 10.1016/j.cbd.2024.101243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/27/2024]
Abstract
Pigmentation genes expressed in skin, body muscle and tail of Thai-flag compared with Blue, White and Red varieties of Siamese fighting fish Betta splendens were identified. In total, 22,919 new unigenes were found. Pearson correlation and PCA analysis revealed that expression profiles of genes in muscle, skin and tail across solid color variety were similar. In contrast, those in skin and red tail part of Thai-flag were closely related but they showed different expression profiles with the white tail part. Moreover, 21,347-64,965 SNPs were identified in exonic regions of identified genes. In total, 28,899 genes were differentially expressed between paired comparisons of libraries where 13,907 genes (48.12 %) were upregulated and 14,992 genes (51.88 %) were downregulated. DEGs between paired libraries were 106-5775 genes relative to the compared libraries (56-2982 and 50-2782 for upregulated and downregulated DEGs). Interestingly, 432 pigmentation genes of B. splendens were found. Of these, 297 DEGs showed differential expression between varieties. Many DEGs in melanogenesis (Bsmcr1r, Bsmcr5r, and Bsslc2a15b), tyrosine metabolism (Bstyr, Bstyrp1b and Bsdct), stripe repressor (BsAsip1 and BsAsip2b), pteridine (Bsgch2) and carotenoid (BsBco2) biosynthesis were downregulated in the Thai-flag compared with solid color varieties. Expression of Bsbco1l, Bsfrem2b, Bskcnj13, Bszic2a and Bspah in skin, muscle and tail of Thai-flag, Blue, Red and White varieties was analyzed by qRT-PCR and revealed differential expression between fish varieties and showed anatomical tissue-preferred expression patterns in the same fish variety. The information could be applied to assist genetic-based development of new B. splendens varieties in the future.
Collapse
Affiliation(s)
- Sureerat Tang
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Sirithorn Janpoom
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Sirikan Prasertlux
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Puttawan Rongmung
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Wanwipa Ittarat
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Onchuda Ratdee
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Bavornlak Khamnamtong
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand
| | - Sirawut Klinbunga
- Aquatic Molecular Genetics and Biotechnology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Khlong Nueng, Khlong Luang, Pathum Thani 12120, Thailand.
| |
Collapse
|
27
|
Suppermpool A, Lyons DG, Broom E, Rihel J. Sleep pressure modulates single-neuron synapse number in zebrafish. Nature 2024; 629:639-645. [PMID: 38693264 PMCID: PMC11096099 DOI: 10.1038/s41586-024-07367-3] [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: 11/14/2022] [Accepted: 03/27/2024] [Indexed: 05/03/2024]
Abstract
Sleep is a nearly universal behaviour with unclear functions1. The synaptic homeostasis hypothesis proposes that sleep is required to renormalize the increases in synaptic number and strength that occur during wakefulness2. Some studies examining either large neuronal populations3 or small patches of dendrites4 have found evidence consistent with the synaptic homeostasis hypothesis, but whether sleep merely functions as a permissive state or actively promotes synaptic downregulation at the scale of whole neurons is unclear. Here, by repeatedly imaging all excitatory synapses on single neurons across sleep-wake states of zebrafish larvae, we show that synapses are gained during periods of wake (either spontaneous or forced) and lost during sleep in a neuron-subtype-dependent manner. However, synapse loss is greatest during sleep associated with high sleep pressure after prolonged wakefulness, and lowest in the latter half of an undisrupted night. Conversely, sleep induced pharmacologically during periods of low sleep pressure is insufficient to trigger synapse loss unless adenosine levels are boosted while noradrenergic tone is inhibited. We conclude that sleep-dependent synapse loss is regulated by sleep pressure at the level of the single neuron and that not all sleep periods are equally capable of fulfilling the functions of synaptic homeostasis.
Collapse
Affiliation(s)
- Anya Suppermpool
- Department of Cell and Developmental Biology, University College London, London, UK
- UCL Ear Institute, University College London, London, UK
| | - Declan G Lyons
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Elizabeth Broom
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, UK.
| |
Collapse
|
28
|
Zaupa M, Nagaraj N, Sylenko A, Baier H, Sawamiphak S, Filosa A. The Calmodulin-interacting peptide Pcp4a regulates feeding state-dependent behavioral choice in zebrafish. Neuron 2024; 112:1150-1164.e6. [PMID: 38295792 DOI: 10.1016/j.neuron.2024.01.001] [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: 03/01/2023] [Revised: 10/06/2023] [Accepted: 01/02/2024] [Indexed: 04/06/2024]
Abstract
Animals constantly need to judge the valence of an object in their environment: is it potential food or a threat? The brain makes fundamental decisions on the appropriate behavioral strategy by integrating external information from sensory organs and internal signals related to physiological needs. For example, a hungry animal may take more risks than a satiated one when deciding to approach or avoid an object. Using a proteomic profiling approach, we identified the Calmodulin-interacting peptide Pcp4a as a key regulator of foraging-related decisions. Food intake reduced abundance of protein and mRNA of pcp4a via dopamine D2-like receptor-mediated repression of adenylate cyclase. Accordingly, deleting the pcp4a gene made zebrafish larvae more risk averse in a binary decision assay. Strikingly, neurons in the tectum became less responsive to prey-like visual stimuli in pcp4a mutants, thus biasing the behavior toward avoidance. This study pinpoints a molecular mechanism modulating behavioral choice according to internal state.
Collapse
Affiliation(s)
- Margherita Zaupa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13092 Berlin, Germany; Freie Universität Berlin, Institute for Biology, 14195 Berlin, Germany
| | - Nagarjuna Nagaraj
- Biochemistry Core Facility, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Anna Sylenko
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13092 Berlin, Germany; Freie Universität Berlin, Institute for Biology, 14195 Berlin, Germany
| | - Herwig Baier
- Max Planck Institute for Biological Intelligence, 82152 Martinsried, Germany
| | - Suphansa Sawamiphak
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13092 Berlin, Germany
| | - Alessandro Filosa
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13092 Berlin, Germany.
| |
Collapse
|
29
|
Greenspan LJ, Ameyaw KK, Castranova D, Mertus CA, Weinstein BM. Live Imaging of Cutaneous Wound Healing after Rotary Tool Injury in Zebrafish. J Invest Dermatol 2024; 144:888-897.e6. [PMID: 37979772 PMCID: PMC10960721 DOI: 10.1016/j.jid.2023.10.015] [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: 12/22/2022] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 11/20/2023]
Abstract
Cutaneous wounds are common afflictions that follow a stereotypical healing process involving hemostasis, inflammation, proliferation, and remodeling phases. In the elderly and those suffering from vascular or metabolic diseases, poor healing after cutaneous injuries can lead to open chronic wounds susceptible to infection. The discovery of new therapeutic strategies to improve this defective wound healing requires a better understanding of the cellular behaviors and molecular mechanisms that drive the different phases of wound healing and how these are altered with age or disease. The zebrafish provides an ideal model for visualization and experimental manipulation of the cellular and molecular events during wound healing in the context of an intact, living vertebrate. To facilitate studies of cutaneous wound healing in zebrafish, we have developed an inexpensive, simple, and effective method for generating reproducible cutaneous injuries in adult zebrafish using a rotary tool. We demonstrate that our injury system can be used in combination with high-resolution live imaging to monitor skin re-epithelialization, immune cell recruitment and activation, and vessel regrowth in the same animal over time. This injury system provides a valuable experimental platform to study key cellular and molecular events during wound healing in vivo with unprecedented resolution.
Collapse
Affiliation(s)
- Leah J Greenspan
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Keith K Ameyaw
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Castranova
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Caleb A Mertus
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
30
|
Jiang K, Xu C, Yu H, Kong L, Liu S, Li Q. Transcriptomic and Physiological Analysis Reveal Melanin Synthesis-Related Genes and Pathways in Pacific Oysters (Crassostrea gigas). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:364-379. [PMID: 38483671 DOI: 10.1007/s10126-024-10302-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 03/06/2024] [Indexed: 04/25/2024]
Abstract
Shell color is one of the shell traits of molluscs, which has been regarded as an economic trait in some bivalves. Pacific oysters (Crassostrea gigas) are important aquaculture shellfish worldwide. In the past decade, several shell color strains of C. gigas were developed through selective breeding, which provides valuable materials for research on the inheritance pattern and regulation mechanisms of shell color. The inheritance patterns of different shell colors in C. gigas have been identified in certain research; however, the regulation mechanism of oyster pigmentation and shell color formation remains unclear. In this study, we performed transcriptomic and physiological analyses using black and white shell oysters to investigate the molecular mechanism of melanin synthesis in C. gigas. Several pigmentation-related pathways, such as cytochrome P450, melanogenesis, tyrosine metabolism, and the cAMP signaling pathway were found. The majority of differentially expressed genes and some signaling molecules from these pathways exhibited a higher level in the black shell oysters than in the white, especially after L-tyrosine feeding, suggesting that those differences may cause a variation of tyrosine metabolism and melanin synthesis. In addition, the in vitro assay using primary cells from mantle tissue showed that L-tyrosine incubation increased cAMP level, gene and protein expression, and melanin content. This study reveals the difference in tyrosine metabolism and melanin synthesis in black and white shell oysters and provides evidence for the potential regulatory mechanism of shell color in oysters.
Collapse
Affiliation(s)
- Kunyin Jiang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Chengxun Xu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Hong Yu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Lingfeng Kong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Shikai Liu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China.
| |
Collapse
|
31
|
Ran R, Li L, Xu T, Huang J, He H, Chen Y. Revealing mitf functions and visualizing allografted tumor metastasis in colorless and immunodeficient Xenopus tropicalis. Commun Biol 2024; 7:275. [PMID: 38443437 PMCID: PMC10915148 DOI: 10.1038/s42003-024-05967-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Transparent immunodeficient animal models not only enhance in vivo imaging investigations of visceral organ development but also facilitate in vivo tracking of transplanted tumor cells. However, at present, transparent and immunodeficient animal models are confined to zebrafish, presenting substantial challenges for real-time, in vivo imaging studies addressing specific biological inquiries. Here, we employed a mitf-/-/prkdc-/-/il2rg-/- triple-knockout strategy to establish a colorless and immunodeficient amphibian model of Xenopus tropicalis. By disrupting the mitf gene, we observed the loss of melanophores, xanthophores, and granular glands in Xenopus tropicalis. Through the endogenous mitf promoter to drive BRAFV600E expression, we confirmed mitf expression in melanophores, xanthophores and granular glands. Moreover, the reconstruction of the disrupted site effectively reinstated melanophores, xanthophores, and granular glands, further highlighting the crucial role of mitf as a regulator in their development. By crossing mitf-/- frogs with prkdc-/-/il2rg-/- frogs, we generated a mitf-/-/prkdc-/-/il2rg-/- Xenopus tropicalis line, providing a colorless and immunodeficient amphibian model. Utilizing this model, we successfully observed intravital metastases of allotransplanted xanthophoromas and migrations of allotransplanted melanomas. Overall, colorless and immunodeficient Xenopus tropicalis holds great promise as a valuable platform for tumorous and developmental biology research.
Collapse
Affiliation(s)
- Rensen Ran
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China.
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Zhuhai, China.
| | - Lanxin Li
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Tingting Xu
- Fujian Medical University Union Hospital, 350001, Fuzhou, China
| | - Jixuan Huang
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Huanhuan He
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, 519000, Zhuhai, China
| | - Yonglong Chen
- Department of Chemical Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China.
| |
Collapse
|
32
|
Zhu Y, Li Q. Mitf involved in shell pigmentation by activating tyrosinase-mediated melanin synthesis in Pacific oyster (Crassostrea gigas). Gene 2024; 897:148086. [PMID: 38104952 DOI: 10.1016/j.gene.2023.148086] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
Pigmentation is frequently observed in the molluscan shells, whereas the molecular regulation about these shell pigments formation is not clear. The microphthalmia-associated transcription factor (Mitf) is an important transactivator in melanin synthesis in vertebrates. Here, the Mitf containing a highly conserved basic helix-loop-helixleucine zipper (bHLH-LZ) domain was identified in an economically important marine bivalve Pacific oyster Crassostrea gigas. The Mitf was found to widespread tissue distribution and the expression was higher in the marginal mantle than in the central mantle. Particularly, the expression level of Mitf was high in black shell color oysters compared with white shell oysters. After injecting siRNA, the expression of Mitf decreased significantly, and the efficiency of RNA interference reached 53%. Besides, knockdown Mitf obviously decreased expression of tyrosinase family genes and tyrosinase activity of mantles, indicating a potential regulatory relationship between Mitf and Tyr or Typs. Simultaneously, there was a sharply reduce in the number of the melanosomes in the outer fold of mantle by silencing of Mitf. Luciferase assays in cell culture further verified that Mitf was involved in transcriptional regulation of Typ-2 and Typ-3 genes through binding to their specific promoter regions. These data argue that Mitf is involved in shell pigmentation through activating tyrosinase-mediated melanin synthesis in C. gigas.
Collapse
Affiliation(s)
- Yijing Zhu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Marine Science Research Institute of Shandong Province (National Oceanographic Center, Qingdao), Qingdao 266104, China
| | - Qi Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
33
|
Schindler M, Endlich N. Zebrafish as a model for podocyte research. Am J Physiol Renal Physiol 2024; 326:F369-F381. [PMID: 38205541 DOI: 10.1152/ajprenal.00335.2023] [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: 10/18/2023] [Revised: 12/25/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Podocytes, specialized postmitotic cells, are central players in various kidney-related diseases. Zebrafish have become a valuable model system for studying podocyte biology because they are genetically easy to manipulate, transparent, and their glomerular structure is similar to that of mammals. This review provides an overview of the knowledge of podocyte biology in zebrafish larvae, with particular focus on their essential contribution to understanding the mechanisms that underlie kidney diseases as well as supporting drug development. In addition, special attention is given to advances in live-imaging techniques allowing the observation of dynamic processes, including podocyte motility, podocyte process behavior, and glomerulus maturation. The review further addresses the functional aspects of podocytes in zebrafish larvae. This includes topics such as glomerular filtration, ultrastructural analyses, and evaluation of podocyte response to nephrotoxic insults. Studies presented in this context have provided important insights into the maintenance and resistance of the glomerular filtration barrier in zebrafish larvae and explored the potential transferability of these findings to mammals such as mice, rats, and most importantly, humans. The recent ability to identify potential therapeutic targets represents a promising new way to identify drugs that could effectively treat podocyte-associated glomerulopathies in humans. In summary, this review gives an overview about the importance of zebrafish as a model for podocyte-related disease and targeted drug development. It also highlights the key role of advanced imaging techniques in transparent zebrafish larvae, improving our understanding of glomerular diseases and the significant potential for translation of these findings to humans.
Collapse
Affiliation(s)
- Maximilian Schindler
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
34
|
Bin JM, Suminaite D, Benito-Kwiecinski SK, Kegel L, Rubio-Brotons M, Early JJ, Soong D, Livesey MR, Poole RJ, Lyons DA. Importin 13-dependent axon diameter growth regulates conduction speeds along myelinated CNS axons. Nat Commun 2024; 15:1790. [PMID: 38413580 PMCID: PMC10899189 DOI: 10.1038/s41467-024-45908-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Axon diameter influences the conduction properties of myelinated axons, both directly, and indirectly through effects on myelin. However, we have limited understanding of mechanisms controlling axon diameter growth in the central nervous system, preventing systematic dissection of how manipulating diameter affects myelination and conduction along individual axons. Here we establish zebrafish to study axon diameter. We find that importin 13b is required for axon diameter growth, but does not affect cell body size or axon length. Using neuron-specific ipo13b mutants, we assess how reduced axon diameter affects myelination and conduction, and find no changes to myelin thickness, precision of action potential propagation, or ability to sustain high frequency firing. However, increases in conduction speed that occur along single myelinated axons with development are tightly linked to their growth in diameter. This suggests that axon diameter growth is a major driver of increases in conduction speeds along myelinated axons over time.
Collapse
Affiliation(s)
- Jenea M Bin
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK.
| | - Daumante Suminaite
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | | | - Linde Kegel
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Maria Rubio-Brotons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Jason J Early
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Daniel Soong
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Matthew R Livesey
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Richard J Poole
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - David A Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
35
|
Huppertz MC, Wilhelm J, Grenier V, Schneider MW, Falt T, Porzberg N, Hausmann D, Hoffmann DC, Hai L, Tarnawski M, Pino G, Slanchev K, Kolb I, Acuna C, Fenk LM, Baier H, Hiblot J, Johnsson K. Recording physiological history of cells with chemical labeling. Science 2024; 383:890-897. [PMID: 38386755 DOI: 10.1126/science.adg0812] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/22/2024] [Indexed: 02/24/2024]
Abstract
Recordings of the physiological history of cells provide insights into biological processes, yet obtaining such recordings is a challenge. To address this, we introduce a method to record transient cellular events for later analysis. We designed proteins that become labeled in the presence of both a specific cellular activity and a fluorescent substrate. The recording period is set by the presence of the substrate, whereas the cellular activity controls the degree of the labeling. The use of distinguishable substrates enabled the recording of successive periods of activity. We recorded protein-protein interactions, G protein-coupled receptor activation, and increases in intracellular calcium. Recordings of elevated calcium levels allowed selections of cells from heterogeneous populations for transcriptomic analysis and tracking of neuronal activities in flies and zebrafish.
Collapse
Affiliation(s)
- Magnus-Carsten Huppertz
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Jonas Wilhelm
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Vincent Grenier
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Martin W Schneider
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Tjalda Falt
- Active Sensing, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Nicola Porzberg
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - David Hausmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk C Hoffmann
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Ling Hai
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Neurology and Neurooncology Program, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Miroslaw Tarnawski
- Protein Expression and Characterization Facility, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Gabriela Pino
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Krasimir Slanchev
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Ilya Kolb
- GENIE Project Team, Janelia Research Campus, Ashburn, VA 20147, USA
| | - Claudio Acuna
- Chica and Heinz Schaller Foundation, Institute of Anatomy and Cell Biology, Heidelberg University, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | - Lisa M Fenk
- Active Sensing, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Herwig Baier
- Department Genes - Circuits - Behavior, Max Planck Institute for Biological Intelligence, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Julien Hiblot
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Kai Johnsson
- Department of Chemical Biology, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
- Institute of Chemical Sciences and Engineering (ISIC), École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
36
|
Tzika AC. On the role of TFEC in reptilian coloration. Front Cell Dev Biol 2024; 12:1358828. [PMID: 38385026 PMCID: PMC10879265 DOI: 10.3389/fcell.2024.1358828] [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: 12/20/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Reptilian species, particularly snakes and lizards, are emerging models of animal coloration. Here, I focus on the role of the TFEC transcription factor in snake and lizard coloration based on a study on wild-type and piebald ball pythons. Genomic mapping previously identified a TFEC mutation linked to the piebald ball python phenotype. The association of TFEC with skin coloration was further supported by gene-editing experiments in the brown anole lizard. However, novel histological analyses presented here reveal discrepancies between the ball python and the anole TFEC mutants phenotype, cautioning against broad generalizations. Indeed, both wild-type and piebald ball pythons completely lack iridophores, whereas the TFEC anole lizard mutants lose their iridophores compared to the wild-type anole. Based on these findings, I discuss the potential role of the MiT/TFE family in skin pigmentation across vertebrate lineages and advocate the need for developmental analyses and additional gene-editing experiments to explore the reptilian coloration diversity.
Collapse
Affiliation(s)
- Athanasia C. Tzika
- Laboratory of Artificial and Natural Evolution (LANE), Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| |
Collapse
|
37
|
Montal E, Suresh S, Ma Y, Tagore MM, White RM. Cancer Modeling by Transgene Electroporation in Adult Zebrafish (TEAZ). Methods Mol Biol 2024; 2707:83-97. [PMID: 37668906 DOI: 10.1007/978-1-0716-3401-1_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023]
Abstract
Transgenic expression of genes is a mainstay of cancer modeling in zebrafish. Traditional transgenic techniques rely upon injection into one-cell embryos, but ideally these transgenes would be expressed only in adult somatic tissues. We provide a method to model cancer in adult zebrafish in which transgenes can be expressed via electroporation. Using melanoma as an example, we demonstrate the feasibility of expressing oncogenes such as BRAFV600E as well as CRISPR/Cas9 inactivation of tumor suppressors such as PTEN. These approaches can be performed in any genetic background such as existing fluorophore reporter lines or the casper line. These methods can readily be extended to other cell types allowing for rapid adult modeling of cancer in zebrafish.
Collapse
Affiliation(s)
- Emily Montal
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shruthy Suresh
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yilun Ma
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohita M Tagore
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard M White
- Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
38
|
Rayamajhi D, Ege M, Ukhanov K, Ringers C, Zhang Y, Jung I, D’Gama PP, Li SS, Cosacak MI, Kizil C, Park HC, Yaksi E, Martens JR, Brody SL, Jurisch-Yaksi N, Roy S. The forkhead transcription factor Foxj1 controls vertebrate olfactory cilia biogenesis and sensory neuron differentiation. PLoS Biol 2024; 22:e3002468. [PMID: 38271330 PMCID: PMC10810531 DOI: 10.1371/journal.pbio.3002468] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024] Open
Abstract
In vertebrates, olfactory receptors localize on multiple cilia elaborated on dendritic knobs of olfactory sensory neurons (OSNs). Although olfactory cilia dysfunction can cause anosmia, how their differentiation is programmed at the transcriptional level has remained largely unexplored. We discovered in zebrafish and mice that Foxj1, a forkhead domain-containing transcription factor traditionally linked with motile cilia biogenesis, is expressed in OSNs and required for olfactory epithelium (OE) formation. In keeping with the immotile nature of olfactory cilia, we observed that ciliary motility genes are repressed in zebrafish, mouse, and human OSNs. Strikingly, we also found that besides ciliogenesis, Foxj1 controls the differentiation of the OSNs themselves by regulating their cell type-specific gene expression, such as that of olfactory marker protein (omp) involved in odor-evoked signal transduction. In line with this, response to bile acids, odors detected by OMP-positive OSNs, was significantly diminished in foxj1 mutant zebrafish. Taken together, our findings establish how the canonical Foxj1-mediated motile ciliogenic transcriptional program has been repurposed for the biogenesis of immotile olfactory cilia, as well as for the development of the OSNs.
Collapse
Affiliation(s)
- Dheeraj Rayamajhi
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Mert Ege
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kirill Ukhanov
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Christa Ringers
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yiliu Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Inyoung Jung
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biomedical Sciences, Korea University, Ansan, South Korea
| | - Percival P. D’Gama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Summer Shijia Li
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Dresden, Germany
| | - Caghan Kizil
- Department of Neurology and The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University, Ansan, South Korea
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
- Koç University Research Center for Translational Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Jeffrey R. Martens
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Steven L. Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
- Department of Paediatrics, National University of Singapore, Singapore
| |
Collapse
|
39
|
Uribe-Arias A, Rozenblat R, Vinepinsky E, Marachlian E, Kulkarni A, Zada D, Privat M, Topsakalian D, Charpy S, Candat V, Nourin S, Appelbaum L, Sumbre G. Radial astrocyte synchronization modulates the visual system during behavioral-state transitions. Neuron 2023; 111:4040-4057.e6. [PMID: 37863038 PMCID: PMC10783638 DOI: 10.1016/j.neuron.2023.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/01/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Glial cells support the function of neurons. Recent evidence shows that astrocytes are also involved in brain computations. To explore whether and how their excitable nature affects brain computations and motor behaviors, we used two-photon Ca2+ imaging of zebrafish larvae expressing GCaMP in both neurons and radial astrocytes (RAs). We found that in the optic tectum, RAs synchronize their Ca2+ transients immediately after the end of an escape behavior. Using optogenetics, ablations, and a genetically encoded norepinephrine sensor, we observed that RA synchronous Ca2+ events are mediated by the locus coeruleus (LC)-norepinephrine circuit. RA synchronization did not induce direct excitation or inhibition of tectal neurons. Nevertheless, it modulated the direction selectivity and the long-distance functional correlations among neurons. This mechanism supports freezing behavior following a switch to an alerted state. These results show that LC-mediated neuro-glial interactions modulate the visual system during transitions between behavioral states.
Collapse
Affiliation(s)
- Alejandro Uribe-Arias
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Rotem Rozenblat
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Ehud Vinepinsky
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Emiliano Marachlian
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Anirudh Kulkarni
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - David Zada
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Martin Privat
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Diego Topsakalian
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sarah Charpy
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Virginie Candat
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Sarah Nourin
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Lior Appelbaum
- The Faculty of Life Sciences and The Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan, Israel
| | - Germán Sumbre
- Institut de Biologie de l'ENS (IBENS), Département de biologie, École normale supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France.
| |
Collapse
|
40
|
Ma L, Zhu Z, Zhang S, Yang R, Liu C, Yu Y, Yang X. Comparative Transcriptome Analysis of the Skin and the Peritoneal Wall Layer of Triplophysa stenura Distributed in High Elevations. BIOLOGY 2023; 13:5. [PMID: 38275726 PMCID: PMC10812932 DOI: 10.3390/biology13010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 01/27/2024]
Abstract
A total of 81,868 All-Unigenes were sequenced and assembled by the transcriptome in the dorsal skin, the lateral skin, and the peritoneal wall layer of Triplophysa stenura with a total assembly length of 123,827,585 bp, and 68,750 unigenes were annotated to seven functional databases. A total of 588 DEGs were screened between the dorsal and lateral skin, 17,097 DEGs were screened between the dorsal skin and the peritoneal wall layer, and 16,598 DEGs were screened between the lateral skin and the peritoneal wall layer. Most of DEGs in three tissues were annotated to GO terms related to cellular structures, binding, cellular processes, and catalytic activity. They were also annotated to KEGG pathways such as the MAPK signaling pathway, PI3K-Akt signaling pathway, Wnt signaling pathway, melanogenesis, tyrosine metabolism, and cell cycle. A total of twenty-three DEGs were found to be enriched in the melanin synthesis pathway by a local Blast comparison, of which nine DEGs were significantly upregulated in the peritoneal wall layer and six DEGs were significantly upregulated in the dorsal and lateral skin. The results suggest that these genes may be associated with the molecular mechanism of melanin synthesis in T. stenura, and the differential regulation of genes may be related to the differences in UVR intensity and tissue sites of melanin synthesis. Further investigation is needed on how these genes specifically regulate melanin synthesis.
Collapse
Affiliation(s)
- Li Ma
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.M.); (Z.Z.); (R.Y.); (C.L.); (Y.Y.)
| | - Zhen Zhu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.M.); (Z.Z.); (R.Y.); (C.L.); (Y.Y.)
- Hubei Vocational College of Bio-Technology, Wuhan 430070, China
| | - Shanzhong Zhang
- Hechuan Campus, Sichuan Fisheries School, Hechuan, Chongqing 401520, China;
| | - Ruibin Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.M.); (Z.Z.); (R.Y.); (C.L.); (Y.Y.)
| | - Chen Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.M.); (Z.Z.); (R.Y.); (C.L.); (Y.Y.)
| | - Yongyao Yu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.M.); (Z.Z.); (R.Y.); (C.L.); (Y.Y.)
| | - Xuefen Yang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; (L.M.); (Z.Z.); (R.Y.); (C.L.); (Y.Y.)
| |
Collapse
|
41
|
Ali MA, Lischka K, Preuss SJ, Trivedi CA, Bollmann JH. A synaptic corollary discharge signal suppresses midbrain visual processing during saccade-like locomotion. Nat Commun 2023; 14:7592. [PMID: 37996414 PMCID: PMC10667368 DOI: 10.1038/s41467-023-43255-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 11/03/2023] [Indexed: 11/25/2023] Open
Abstract
In motor control, the brain not only sends motor commands to the periphery, but also generates concurrent internal signals known as corollary discharge (CD) that influence sensory information processing around the time of movement. CD signals are important for identifying sensory input arising from self-motion and to compensate for it, but the underlying mechanisms remain unclear. Using whole-cell patch clamp recordings from neurons in the zebrafish optic tectum, we discovered an inhibitory synaptic signal, temporally locked to spontaneous and visually driven locomotion. This motor-related inhibition was appropriately timed to counteract visually driven excitatory input arising from the fish's own motion, and transiently suppressed tectal spiking activity. High-resolution calcium imaging revealed localized motor-related signals in the tectal neuropil and the upstream torus longitudinalis, suggesting that CD enters the tectum via this pathway. Together, our results show how visual processing is suppressed during self-motion by motor-related phasic inhibition. This may help explain perceptual saccadic suppression observed in many species.
Collapse
Affiliation(s)
- Mir Ahsan Ali
- Developmental Biology, Institute of Biology I, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Katharina Lischka
- Developmental Biology, Institute of Biology I, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Stephanie J Preuss
- Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Springer Nature Group, Heidelberg, Germany
| | - Chintan A Trivedi
- Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Dept Cell and Developmental Biology, University College London, London, UK
| | - Johann H Bollmann
- Developmental Biology, Institute of Biology I, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.
- Max Planck Institute for Medical Research, 69120, Heidelberg, Germany.
- Bernstein Center Freiburg, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
42
|
Bian WP, Xie SL, Wang C, Martinovich GG, Ma YB, Jia PP, Pei DS. mitfa deficiency promotes immune vigor and potentiates antitumor effects in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109130. [PMID: 37777099 DOI: 10.1016/j.fsi.2023.109130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
The mitfa gene is a well-known transcription factor associated with microphthalmia and is essential for early melanophore development. However, little is known about how mitfa affects the immune system. Here, we generated a novel mitfa knock-out zebrafish line using the CRISPR/Cas9 system. The mitfa-/- zebrafish exhibited reduced melanin levels compared to the nacre mutant. We investigated the impact on the immune system after exposure to Edwardsiella tarda and bifenazate in zebrafish larvae, and observed that the macrophage numbers were reduced in both treated groups. Remarkably, the expression levels of immune-related genes exhibited significant increases after bacterial challenge or bifenazate exposure in the mitfa-/- zebrafish, except for tlr4 and rela. Furthermore, we conducted xenograft experiments using mouse B16 melanoma cells. Notably, the cancer cells didn't show a high cell migration ratio, implying that the immune system was highly activated after the loss of mifta. Taken together, our findings suggest that mitfa-/- zebrafish serve as a valuable model for investigating the relationship between the immune system and melanocytes, providing new insights into the role of mitfa in immune responses.
Collapse
Affiliation(s)
- Wan-Ping Bian
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Shao-Lin Xie
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Chao Wang
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | | | - Yan-Bo Ma
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Pan-Pan Jia
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
43
|
Derudas M, O’Reilly M, Kirkwood NK, Kenyon EJ, Grimsey S, Kitcher SR, Workman S, Bull JC, Ward SE, Kros CJ, Richardson GP. Charge and lipophilicity are required for effective block of the hair-cell mechano-electrical transducer channel by FM1-43 and its derivatives. Front Cell Dev Biol 2023; 11:1247324. [PMID: 37900280 PMCID: PMC10601989 DOI: 10.3389/fcell.2023.1247324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
The styryl dye FM1-43 is widely used to study endocytosis but behaves as a permeant blocker of the mechano-electrical transducer (MET) channel in sensory hair cells, loading rapidly and specifically into the cytoplasm of hair cells in a MET channel-dependent manner. Patch clamp recordings of mouse outer hair cells (OHCs) were used to determine how a series of structural modifications of FM1-43 affect MET channel block. Fluorescence microscopy was used to assess how the modifications influence hair-cell loading in mouse cochlear cultures and zebrafish neuromasts. Cochlear cultures were also used to evaluate otoprotective potential of the modified FM1-43 derivatives. Structure-activity relationships reveal that the lipophilic tail and the cationic head group of FM1-43 are both required for MET channel block in mouse cochlear OHCs; neither moiety alone is sufficient. The extent of MET channel block is augmented by increasing the lipophilicity/bulkiness of the tail, by reducing the number of positive charges in the head group from two to one, or by increasing the distance between the two charged head groups. Loading assays with zebrafish neuromasts and mouse cochlear cultures are broadly in accordance with these observations but reveal a loss of hair-cell specific labelling with increasing lipophilicity. Although FM1-43 and many of its derivatives are generally cytotoxic when tested on cochlear cultures in the presence of an equimolar concentration of the ototoxic antibiotic gentamicin (5 µM), at a 10-fold lower concentration (0.5 µM), two of the derivatives protect OHCs from cell death caused by 48 h-exposure to 5 µM gentamicin.
Collapse
Affiliation(s)
- Marco Derudas
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Molly O’Reilly
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, Netherlands
| | - Nerissa K. Kirkwood
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Emma J. Kenyon
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- School of Medicine, Institute of Life Sciences, Swansea University, Swansea, United Kingdom
| | - Sybil Grimsey
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Siân R. Kitcher
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Section on Neuronal Circuitry, National Institute on Deafness and Other Communication Disorders NIH, Bethesda, MD, United States
| | - Shawna Workman
- Department of Biosciences, College of Science, Swansea University, Swansea, United Kingdom
| | - James C. Bull
- Department of Biosciences, College of Science, Swansea University, Swansea, United Kingdom
| | - Simon E. Ward
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Medicines Discovery Institute, Cardiff University, Cardiff, United Kingdom
| | - Corné J. Kros
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Guy P. Richardson
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
44
|
Asad A, Shahidan NO, de la Vega de León A, Wiggin GR, Whitfield TT, Baxendale S. A screen of pharmacologically active compounds to identify modulators of the Adgrg6/Gpr126 signalling pathway in zebrafish embryos. Basic Clin Pharmacol Toxicol 2023; 133:364-377. [PMID: 37394692 PMCID: PMC10952222 DOI: 10.1111/bcpt.13923] [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: 04/06/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
Adhesion G protein-coupled receptors (GPCRs) are an underrepresented class of GPCRs in drug discovery. We previously developed an in vivo drug screening pipeline to identify compounds with agonist activity for Adgrg6 (Gpr126), an adhesion GPCR required for myelination of the peripheral nervous system in vertebrates. The screening assay tests for rescue of an ear defect found in adgrg6tb233c-/- hypomorphic homozygous mutant zebrafish, using the expression of versican b (vcanb) mRNA as an easily identifiable phenotype. In the current study, we used the same assay to screen a commercially available library of 1280 diverse bioactive compounds (Sigma LOPAC). Comparison with published hits from two partially overlapping compound collections (Spectrum, Tocris) confirms that the screening assay is robust and reproducible. Using a modified counter screen for myelin basic protein (mbp) gene expression, we have identified 17 LOPAC compounds that can rescue both inner ear and myelination defects in adgrg6tb233c-/- hypomorphic mutants, three of which (ebastine, S-methylisothiourea hemisulfate, and thapsigargin) are new hits. A further 25 LOPAC hit compounds were effective at rescuing the otic vcanb expression but not mbp. Together, these and previously identified hits provide a wealth of starting material for the development of novel and specific pharmacological modulators of Adgrg6 receptor activity.
Collapse
Affiliation(s)
- Anzar Asad
- School of BiosciencesUniversity of SheffieldSheffieldUK
| | | | | | | | | | - Sarah Baxendale
- School of BiosciencesUniversity of SheffieldSheffieldUK
- Sheffield Zebrafish Screening Facility, School of BiosciencesUniversity of SheffieldSheffieldUK
| |
Collapse
|
45
|
Miyadai M, Takada H, Shiraishi A, Kimura T, Watakabe I, Kobayashi H, Nagao Y, Naruse K, Higashijima SI, Shimizu T, Kelsh RN, Hibi M, Hashimoto H. A gene regulatory network combining Pax3/7, Sox10 and Mitf generates diverse pigment cell types in medaka and zebrafish. Development 2023; 150:dev202114. [PMID: 37823232 PMCID: PMC10617610 DOI: 10.1242/dev.202114] [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: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023]
Abstract
Neural crest cells generate numerous derivatives, including pigment cells, and are a model for studying how fate specification from multipotent progenitors is controlled. In mammals, the core gene regulatory network for melanocytes (their only pigment cell type) contains three transcription factors, Sox10, Pax3 and Mitf, with the latter considered a master regulator of melanocyte development. In teleosts, which have three to four pigment cell types (melanophores, iridophores and xanthophores, plus leucophores e.g. in medaka), gene regulatory networks governing fate specification are poorly understood, although Mitf function is considered conserved. Here, we show that the regulatory relationships between Sox10, Pax3 and Mitf are conserved in zebrafish, but the role for Mitf is more complex than previously emphasized, affecting xanthophore development too. Similarly, medaka Mitf is necessary for melanophore, xanthophore and leucophore formation. Furthermore, expression patterns and mutant phenotypes of pax3 and pax7 suggest that Pax3 and Pax7 act sequentially, activating mitf expression. Pax7 modulates Mitf function, driving co-expressing cells to differentiate as xanthophores and leucophores rather than melanophores. We propose that pigment cell fate specification should be considered to result from the combinatorial activity of Mitf with other transcription factors.
Collapse
Affiliation(s)
- Motohiro Miyadai
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Hiroyuki Takada
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Akiko Shiraishi
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Tetsuaki Kimura
- Laboratory of Bioresources, National Institute for Basic Biology, Okazaki 444-8585, Japan
| | - Ikuko Watakabe
- National Institutes of Natural Sciences, Exploratory Research Center on Life and Living Systems, National Institute for Basic Biology, Okazaki 444-8787, Japan
| | - Hikaru Kobayashi
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Yusuke Nagao
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Kiyoshi Naruse
- Laboratory of Bioresources, National Institute for Basic Biology, Okazaki 444-8585, Japan
| | - Shin-ichi Higashijima
- National Institutes of Natural Sciences, Exploratory Research Center on Life and Living Systems, National Institute for Basic Biology, Okazaki 444-8787, Japan
| | - Takashi Shimizu
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Robert N. Kelsh
- Department of Life Sciences, University of Bath, Bath BA2 7AY, UK
| | - Masahiko Hibi
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| | - Hisashi Hashimoto
- Laboratory of Biological Science, Division of Natural Science, Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan
| |
Collapse
|
46
|
Martins TG, Soliman R, Cordero-Maldonado ML, Donato C, Ameli C, Mombaerts L, Skupin A, Peri F, Crawford AD. Seizure-induced increase in microglial cell population in the developing zebrafish brain. Epilepsy Res 2023; 195:107203. [PMID: 37572541 DOI: 10.1016/j.eplepsyres.2023.107203] [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: 03/03/2023] [Revised: 06/21/2023] [Accepted: 07/31/2023] [Indexed: 08/14/2023]
Abstract
Epilepsy is a chronic brain disorder characterized by unprovoked and recurrent seizures, of which 60% are of unknown etiology. Recent studies implicate microglia in the pathophysiology of epilepsy. However, their role in this process, in particular following early-life seizures, remains poorly understood due in part to the lack of suitable experimental models allowing the in vivo imaging of microglial activity. Given the advantage of zebrafish larvae for minimally-invasive imaging approaches, we sought for the first time to describe the microglial responses after acute seizures in two different zebrafish larval models: a chemically-induced epileptic model by the systemic injection of kainate at 3 days post-fertilization, and the didys552 genetic epilepsy model, which carries a mutation in scn1lab that leads to spontaneous epileptiform discharges. Kainate-treated larvae exhibited transient brain damage as shown by increased numbers of apoptotic nuclei as early as one day post-injection, which was followed by an increase in the number of microglia in the brain. A similar microglial phenotype was also observed in didys552-/- mutants, suggesting that microglia numbers change in response to seizure-like activity in the brain. Interestingly, kainate-treated larvae also displayed a decreased seizure threshold towards subsequent pentylenetetrazole-induced seizures, as shown by higher locomotor and encephalographic activity in comparison with vehicle-injected larvae. These results are comparable to kainate-induced rodent seizure models and suggest the suitability of these zebrafish seizure models for future studies, in particular to elucidate the links between epileptogenesis and microglial dynamic changes after seizure induction in the developing brain, and to understand how these modulate seizure susceptibility.
Collapse
Affiliation(s)
- Teresa G Martins
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Remon Soliman
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | - Cristina Donato
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Corrado Ameli
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Laurent Mombaerts
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Francesca Peri
- Developmental Biology Group, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Institute for Orphan Drug Discovery, Bremerhaven, Germany.
| |
Collapse
|
47
|
Hagio H, Koyama W, Hosaka S, Song AD, Narantsatsral J, Matsuda K, Sugihara T, Shimizu T, Koyanagi M, Terakita A, Hibi M. Optogenetic manipulation of Gq- and Gi/o-coupled receptor signaling in neurons and heart muscle cells. eLife 2023; 12:e83974. [PMID: 37589544 PMCID: PMC10435233 DOI: 10.7554/elife.83974] [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: 10/05/2022] [Accepted: 07/27/2023] [Indexed: 08/18/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) transmit signals into cells depending on the G protein type. To analyze the functions of GPCR signaling, we assessed the effectiveness of animal G-protein-coupled bistable rhodopsins that can be controlled into active and inactive states by light application using zebrafish. We expressed Gq- and Gi/o-coupled bistable rhodopsins in hindbrain reticulospinal V2a neurons, which are involved in locomotion, or in cardiomyocytes. Light stimulation of the reticulospinal V2a neurons expressing Gq-coupled spider Rh1 resulted in an increase in the intracellular Ca2+ level and evoked swimming behavior. Light stimulation of cardiomyocytes expressing the Gi/o-coupled mosquito Opn3, pufferfish TMT opsin, or lamprey parapinopsin induced cardiac arrest, and the effect was suppressed by treatment with pertussis toxin or barium, suggesting that Gi/o-dependent regulation of inward-rectifier K+ channels controls cardiac function. These data indicate that these rhodopsins are useful for optogenetic control of GPCR-mediated signaling in zebrafish neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Hanako Hagio
- Graduate School of Science, Nagoya UniversityNagoyaJapan
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Institute for Advanced Research, Nagoya UniversityNagoyaJapan
| | - Wataru Koyama
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | - Shiori Hosaka
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | | | | | - Koji Matsuda
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| | | | | | | | - Akihisa Terakita
- Graduate School of Science, Osaka Metropolitan UniversityOsakaJapan
| | - Masahiko Hibi
- Graduate School of Science, Nagoya UniversityNagoyaJapan
| |
Collapse
|
48
|
Hagio H, Koyama W, Hosaka S, Song AD, Narantsatsral J, Matsuda K, Shimizu T, Hososhima S, Tsunoda SP, Kandori H, Hibi M. Optogenetic manipulation of neuronal and cardiomyocyte functions in zebrafish using microbial rhodopsins and adenylyl cyclases. eLife 2023; 12:e83975. [PMID: 37589546 PMCID: PMC10435232 DOI: 10.7554/elife.83975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 07/25/2023] [Indexed: 08/18/2023] Open
Abstract
Even though microbial photosensitive proteins have been used for optogenetics, their use should be optimized to precisely control cell and tissue functions in vivo. We exploited GtCCR4 and KnChR, cation channelrhodopsins from algae, BeGC1, a guanylyl cyclase rhodopsin from a fungus, and photoactivated adenylyl cyclases (PACs) from cyanobacteria (OaPAC) or bacteria (bPAC), to control cell functions in zebrafish. Optical activation of GtCCR4 and KnChR in the hindbrain reticulospinal V2a neurons, which are involved in locomotion, induced swimming behavior at relatively short latencies, whereas activation of BeGC1 or PACs achieved it at long latencies. Activation of GtCCR4 and KnChR in cardiomyocytes induced cardiac arrest, whereas activation of bPAC gradually induced bradycardia. KnChR activation led to an increase in intracellular Ca2+ in the heart, suggesting that depolarization caused cardiac arrest. These data suggest that these optogenetic tools can be used to reveal the function and regulation of zebrafish neurons and cardiomyocytes.
Collapse
Affiliation(s)
- Hanako Hagio
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
- Graduate School of Bioagricultural Sciences, Nagoya UniversityNagoyaJapan
- Institute for Advanced Research, Nagoya UniversityNagoyaJapan
| | - Wataru Koyama
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Shiori Hosaka
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | | | | | - Koji Matsuda
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Takashi Shimizu
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| | - Shoko Hososhima
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Satoshi P Tsunoda
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Hideki Kandori
- Department of Life Science and Applied Chemistry, Nagoya Institute of TechnologyNagoyaJapan
| | - Masahiko Hibi
- Graduate School of Science, Nagoya University, JapanNagoyaJapan
| |
Collapse
|
49
|
Podobnik M, Singh AP, Fu Z, Dooley CM, Frohnhöfer HG, Firlej M, Stednitz SJ, Elhabashy H, Weyand S, Weir JR, Lu J, Nüsslein-Volhard C, Irion U. kcnj13 regulates pigment cell shapes in zebrafish and has diverged by cis-regulatory evolution between Danio species. Development 2023; 150:dev201627. [PMID: 37530080 PMCID: PMC10482006 DOI: 10.1242/dev.201627] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023]
Abstract
Teleost fish of the genus Danio are excellent models to study the genetic and cellular bases of pigment pattern variation in vertebrates. The two sister species Danio rerio and Danio aesculapii show divergent patterns of horizontal stripes and vertical bars that are partly caused by the divergence of the potassium channel gene kcnj13. Here, we show that kcnj13 is required only in melanophores for interactions with xanthophores and iridophores, which cause location-specific pigment cell shapes and thereby influence colour pattern and contrast in D. rerio. Cis-regulatory rather than protein coding changes underlie kcnj13 divergence between the two Danio species. Our results suggest that homotypic and heterotypic interactions between the pigment cells and their shapes diverged between species by quantitative changes in kcnj13 expression during pigment pattern diversification.
Collapse
Affiliation(s)
- Marco Podobnik
- Max Planck Institute for Biology, 72076 Tübingen, Germany
| | - Ajeet P. Singh
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Zhenqiang Fu
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
| | - Christopher M. Dooley
- Department of Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | | | - Magdalena Firlej
- Friedrich Miescher Laboratory of the Max Planck Society, 72076 Tübingen, Germany
| | - Sarah J. Stednitz
- Department of Anatomy & Physiology, University of Melbourne, Victoria, 3010, Melbourne, Australia
| | - Hadeer Elhabashy
- Department of Protein Evolution, Max Planck Institute for Biology, 72076 Tübingen, Germany
- Institute for Bioinformatics and Medical Informatics, University of Tübingen, 72076 Tübingen, Germany
- Department of Computer Science, University of Tübingen, 72076 Tübingen, Germany
| | - Simone Weyand
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | - John R. Weir
- Friedrich Miescher Laboratory of the Max Planck Society, 72076 Tübingen, Germany
| | - Jianguo Lu
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
| | | | - Uwe Irion
- Max Planck Institute for Biology, 72076 Tübingen, Germany
| |
Collapse
|
50
|
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
|