1
|
Markert SM. Studying zebrafish nervous system structure and function in health and disease with electron microscopy. Dev Growth Differ 2023; 65:502-516. [PMID: 37740826 PMCID: PMC11520969 DOI: 10.1111/dgd.12890] [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: 05/31/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 09/25/2023]
Abstract
Zebrafish (Danio rerio) is a well-established model for studying the nervous system. Findings in zebrafish often inform studies on human diseases of the nervous system and provide crucial insight into disease mechanisms. The functions of the nervous system often rely on communication between neurons. Signal transduction is achieved via release of signaling molecules in the form of neuropeptides or neurotransmitters at synapses. Snapshots of membrane dynamics of these processes are imaged by electron microscopy. Electron microscopy can reveal ultrastructure and thus synaptic processes. This is crucial both for mapping synaptic connections and for investigating synaptic functions. In addition, via volumetric electron microscopy, the overall architecture of the nervous system becomes accessible, where structure can inform function. Electron microscopy is thus of particular value for studying the nervous system. However, today a plethora of electron microscopy techniques and protocols exist. Which technique is most suitable highly depends on the research question and scope as well as on the type of tissue that is examined. This review gives an overview of the electron microcopy techniques used on the zebrafish nervous system. It aims to give researchers a guide on which techniques are suitable for their specific questions and capabilities as well as an overview of the capabilities of electron microscopy in neurobiological research in the zebrafish model.
Collapse
|
2
|
Lei X, Tian X, Wang H, Xu X, Li G, Liu W, Wang D, Xiao Z, Zhang M, Li MJ, Zhang Z, Ma Z, Liu Z. Noncoding SNP at rs1663689 represses ADGRG6 via interchromosomal interaction and reduces lung cancer progression. EMBO Rep 2023; 24:e56212. [PMID: 37154297 PMCID: PMC10328068 DOI: 10.15252/embr.202256212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 05/10/2023] Open
Abstract
A previous genome-wide association study (GWAS) revealed an association of the noncoding SNP rs1663689 with susceptibility to lung cancer in the Chinese population. However, the underlying mechanism is unknown. In this study, using allele-specific 4C-seq in heterozygous lung cancer cells combined with epigenetic information from CRISPR/Cas9-edited cell lines, we show that the rs1663689 C/C variant represses the expression of ADGRG6, a gene located on a separate chromosome, through an interchromosomal interaction of the rs1663689 bearing region with the ADGRG6 promoter. This reduces downstream cAMP-PKA signaling and subsequently tumor growth both in vitro and in xenograft models. Using patient-derived organoids, we show that rs1663689 T/T-but not C/C-bearing lung tumors are sensitive to the PKA inhibitor H89, potentially informing therapeutic strategies. Our study identifies a genetic variant-mediated interchromosomal interaction underlying ADGRG6 regulation and suggests that targeting the cAMP-PKA signaling pathway may be beneficial in lung cancer patients bearing the homozygous risk genotype at rs1663689.
Collapse
Affiliation(s)
- Xinyue Lei
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Xiaoling Tian
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Hao Wang
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Xinran Xu
- Department of Pharmacology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Guoli Li
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Wenxu Liu
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Dan Wang
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Zengtuan Xiao
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Mengzhe Zhang
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Mulin Jun Li
- Department of Pharmacology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Zhenfa Zhang
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Zhenyi Ma
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Department of Cell Biology, School of Basic Medical SciencesHangzhou Normal UniversityHangzhouChina
| | - Zhe Liu
- Department of Lung Cancer CenterTianjin Medical University Cancer Institute and HospitalHaihe Laboratory of Cell EcosystemState Key Laboratory of Experimental HematologyDepartment of UrologyThe Second Hospital of Tianjin Medical UniversityKey Laboratory of Immune Microenvironment and Disease of the Ministry of EducationDepartment of ImmunologySchool of Basic Medical SciencesTianjin Medical UniversityTianjinChina
- Department of Pharmacology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Department of Cell Biology, School of Basic Medical SciencesHangzhou Normal UniversityHangzhouChina
- Collaborative Innovation Center for Cancer Personalized MedicineNanjing Medical UniversityNanjingChina
| |
Collapse
|
3
|
Role of Primary Cilia in Skeletal Disorders. Stem Cells Int 2022; 2022:6063423. [PMID: 35761830 PMCID: PMC9233574 DOI: 10.1155/2022/6063423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/23/2022] [Accepted: 06/03/2022] [Indexed: 11/26/2022] Open
Abstract
Primary cilia are highly conserved microtubule-based organelles that project from the cell surface into the extracellular environment and play important roles in mechanosensation, mechanotransduction, polarity maintenance, and cell behaviors during organ development and pathological changes. Intraflagellar transport (IFT) proteins are essential for cilium formation and function. The skeletal system consists of bones and connective tissue, including cartilage, tendons, and ligaments, providing support, stability, and movement to the body. Great progress has been achieved in primary cilia and skeletal disorders in recent decades. Increasing evidence suggests that cells with cilium defects in the skeletal system can cause numerous human diseases. Moreover, specific deletion of ciliary proteins in skeletal tissues with different Cre mice resulted in diverse malformations, suggesting that primary cilia are involved in the development of skeletal diseases. In addition, the intact of primary cilium is essential to osteogenic/chondrogenic induction of mesenchymal stem cells, regarded as a promising target for clinical intervention for skeletal disorders. In this review, we summarized the role of primary cilia and ciliary proteins in the pathogenesis of skeletal diseases, including osteoporosis, bone/cartilage tumor, osteoarthritis, intervertebral disc degeneration, spine scoliosis, and other cilium-related skeletal diseases, and highlighted their promising treatment methods, including using mesenchymal stem cells. Our review tries to present evidence for primary cilium as a promising target for clinical intervention for skeletal diseases.
Collapse
|
4
|
Pan C, Zhu F, Wu M, Jiang L, Zhao X, Yang M. Degradation and toxicity of the antidepressant fluoxetine in an aqueous system by UV irradiation. CHEMOSPHERE 2022; 287:132434. [PMID: 34606890 DOI: 10.1016/j.chemosphere.2021.132434] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/05/2021] [Accepted: 09/17/2021] [Indexed: 06/13/2023]
Abstract
Fluoxetine (FLU), a selective serotonin reuptake inhibitor, is commonly found in aquatic environments. Ultraviolet (UV) photolysis is widely used to remove certain pharmaceuticals from water and wastewater. The present study aimed to investigate the toxicity of FLU and its transformed products formed during UV photolysis by using zebrafish embryos (Danio rerio) as a model. The degradation rates of FLU for five days were approximately 63.6% ± 2.14%, 84.6% ± 0.99%, and 97.5% ± 0.25% after 15, 30, and 60 min of UV irradiation, respectively. Furthermore, the degradation mechanism was explored using LC-MS measurements and density flooding theory (DFT) theoretical calculations. Comprehensive toxicity preassessment of FLU and its degradation products was carried out using the T.E.S.T. software. The effects of physiological and biochemical parameters and neuron- and apoptosis-related gene expression were examined in zebrafish embryos exposed to non-irradiated (0-min) and irradiated (15, 30- and 60-min) solutions from 4 h post-fertilization (hpf) to 120 hpf. The hatching time of zebrafish embryos exposed to the non-irradiated solution (0-min) and irradiated solution (60-min) was delayed, their heart rate at 48 and 72 hpf increased, and their body length at 120 hpf decreased. Significant differences were found between the non-irradiated (0-min) and UV-irradiated (15- or 30-min) groups. A dynamic response involving acetylcholinesterase (AChE) and superoxide dismutase (SOD) activity was also observed in the non-irradiated and UV-irradiated groups. During the UV treatment experiments, the expression levels of neuron-related and apoptosis-related genes were significantly reduced over time alongside the formation of FLU degradation products. Overall, this study provides new concepts to remove and assess the toxicity of emerging contaminants in aquatic environments and highlights the need to consider the formation and persistence of toxic transformation products.
Collapse
Affiliation(s)
- Chenyuan Pan
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Feng Zhu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Minghong Wu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Lihui Jiang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Xiaoyu Zhao
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| | - Ming Yang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
5
|
Neely SA, Lyons DA. Insights Into Central Nervous System Glial Cell Formation and Function From Zebrafish. Front Cell Dev Biol 2021; 9:754606. [PMID: 34912801 PMCID: PMC8666443 DOI: 10.3389/fcell.2021.754606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/05/2021] [Indexed: 12/23/2022] Open
Abstract
The term glia describes a heterogenous collection of distinct cell types that make up a large proportion of our nervous system. Although once considered the glue of the nervous system, the study of glial cells has evolved significantly in recent years, with a large body of literature now highlighting their complex and diverse roles in development and throughout life. This progress is due, in part, to advances in animal models in which the molecular and cellular mechanisms of glial cell development and function as well as neuron-glial cell interactions can be directly studied in vivo in real time, in intact neural circuits. In this review we highlight the instrumental role that zebrafish have played as a vertebrate model system for the study of glial cells, and discuss how the experimental advantages of the zebrafish lend themselves to investigate glial cell interactions and diversity. We focus in particular on recent studies that have provided insight into the formation and function of the major glial cell types in the central nervous system in zebrafish.
Collapse
Affiliation(s)
- Sarah A. Neely
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
6
|
Lv X, Xu J, Jiang J, Wu P, Tan R, Wang B. Genetic animal models of scoliosis: A systematical review. Bone 2021; 152:116075. [PMID: 34174503 DOI: 10.1016/j.bone.2021.116075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Scoliosis is a complex disease with undetermined pathogenesis and has a strong relationship with genetics. Models of scoliosis in animals have been established for better comprehending its pathogenesis and treatment. In this review, we searched all the genetic animal models with body curvature in databases, and reviewed the related genes and scoliosis types. Meanwhile, we also summarized the pathogenesis of scoliosis reported so far. Summarizing the positive phenotypic animal models contributes to a better understanding on the pathogenesis of scoliosis and facilitates the selection of experimental models when a possible pathogenic factor is concerned.
Collapse
Affiliation(s)
- Xin Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jinghong Xu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jiajiong Jiang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Pengfei Wu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Renchun Tan
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Bing Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
7
|
Panlilio JM, Jones IT, Salanga MC, Aluru N, Hahn ME. Developmental Exposure to Domoic Acid Disrupts Startle Response Behavior and Circuitry in Zebrafish. Toxicol Sci 2021; 182:310-326. [PMID: 34097058 DOI: 10.1093/toxsci/kfab066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Harmful algal blooms produce potent neurotoxins that accumulate in seafood and are hazardous to human health. Developmental exposure to the harmful algal bloom toxin, domoic acid (DomA), has behavioral consequences well into adulthood, but the cellular and molecular mechanisms of DomA developmental neurotoxicity are largely unknown. To assess these, we exposed zebrafish embryos to DomA during the previously identified window of susceptibility and used the well-known startle response circuit as a tool to identify specific neuronal components that are targeted by exposure to DomA. Exposure to DomA reduced startle responsiveness to both auditory/vibrational and electrical stimuli, and even at the highest stimulus intensities tested, led to a dramatic reduction of one type of startle (short-latency c-starts). Furthermore, DomA-exposed larvae had altered kinematics for both types of startle responses tested, exhibiting shallower bend angles and slower maximal angular velocities. Using vital dye staining, immunolabeling, and live imaging of transgenic lines, we determined that although the sensory inputs were intact, the reticulospinal neurons required for short-latency c-starts were absent in most DomA-exposed larvae. Furthermore, axon tracing revealed that DomA-treated larvae also showed significantly reduced primary motor neuron axon collaterals. Overall, these results show that developmental exposure to DomA targets large reticulospinal neurons and motor neuron axon collaterals, resulting in measurable deficits in startle behavior. They further provide a framework for using the startle response circuit to identify specific neural populations disrupted by toxins or toxicants and to link these disruptions to functional consequences for neural circuit function and behavior.
Collapse
Affiliation(s)
- Jennifer M Panlilio
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.,Massachusetts Institute of Technology (MIT) - Woods Hole Oceanographic Institution (WHOI) Joint Graduate Program in Oceanography and Oceanographic Engineering, Massachusetts 02543, USA.,Woods Hole Center for Oceans and Human Health, Woods Hole, Massachusetts 02543, USA
| | - Ian T Jones
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.,Massachusetts Institute of Technology (MIT) - Woods Hole Oceanographic Institution (WHOI) Joint Graduate Program in Oceanography and Oceanographic Engineering, Massachusetts 02543, USA
| | - Matthew C Salanga
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA
| | - Neelakanteswar Aluru
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.,Woods Hole Center for Oceans and Human Health, Woods Hole, Massachusetts 02543, USA
| | - Mark E Hahn
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, Massachusetts 02543, USA.,Woods Hole Center for Oceans and Human Health, Woods Hole, Massachusetts 02543, USA
| |
Collapse
|
8
|
Siems SB, Jahn O, Hoodless LJ, Jung RB, Hesse D, Möbius W, Czopka T, Werner HB. Proteome Profile of Myelin in the Zebrafish Brain. Front Cell Dev Biol 2021; 9:640169. [PMID: 33898427 PMCID: PMC8060510 DOI: 10.3389/fcell.2021.640169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
The velocity of nerve conduction along vertebrate axons depends on their ensheathment with myelin. Myelin membranes comprise specialized proteins well characterized in mice. Much less is known about the protein composition of myelin in non-mammalian species. Here, we assess the proteome of myelin biochemically purified from the brains of adult zebrafish (Danio rerio), considering its increasing popularity as model organism for myelin biology. Combining gel-based and gel-free proteomic approaches, we identified > 1,000 proteins in purified zebrafish myelin, including all known constituents. By mass spectrometric quantification, the predominant Ig-CAM myelin protein zero (MPZ/P0), myelin basic protein (MBP), and the short-chain dehydrogenase 36K constitute 12%, 8%, and 6% of the total myelin protein, respectively. Comparison with previously established mRNA-abundance profiles shows that expression of many myelin-related transcripts coincides with the maturation of zebrafish oligodendrocytes. Zebrafish myelin comprises several proteins that are not present in mice, including 36K, CLDNK, and ZWI. However, a surprisingly large number of ortholog proteins is present in myelin of both species, indicating partial evolutionary preservation of its constituents. Yet, the relative abundance of CNS myelin proteins can differ markedly as exemplified by the complement inhibitor CD59 that constitutes 5% of the total zebrafish myelin protein but is a low-abundant myelin component in mice. Using novel transgenic reporter constructs and cryo-immuno electron microscopy, we confirm the incorporation of CD59 into myelin sheaths. These data provide the first proteome resource of zebrafish CNS myelin and demonstrate both similarities and heterogeneity of myelin composition between teleost fish and rodents.
Collapse
Affiliation(s)
- Sophie B Siems
- Department of Neurogenetics, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Laura J Hoodless
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Dörte Hesse
- Proteomics Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Tim Czopka
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| |
Collapse
|
9
|
Baxendale S, Asad A, Shahidan NO, Wiggin GR, Whitfield TT. The adhesion GPCR Adgrg6 (Gpr126): Insights from the zebrafish model. Genesis 2021; 59:e23417. [PMID: 33735533 PMCID: PMC11475505 DOI: 10.1002/dvg.23417] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/13/2022]
Abstract
Adhesion GPCRs are important regulators of conserved developmental processes and represent an untapped pool of potential targets for drug discovery. The adhesion GPCR Adgrg6 (Gpr126) has critical developmental roles in Schwann cell maturation and inner ear morphogenesis in the zebrafish embryo. Mutations in the human ADGRG6 gene can result in severe deficits in peripheral myelination, and variants have been associated with many other disease conditions. Here, we review work on the zebrafish Adgrg6 signaling pathway and its potential as a disease model. Recent advances have been made in the analysis of the structure of the Adgrg6 receptor, demonstrating alternative structural conformations and the presence of a conserved calcium-binding site within the CUB domain of the extracellular region that is critical for receptor function. Homozygous zebrafish adgrg6 hypomorphic mutants have been used successfully as a whole-animal screening platform, identifying candidate molecules that can influence signaling activity and rescue mutant phenotypes. These compounds offer promise for further development as small molecule modulators of Adgrg6 pathway activity.
Collapse
Affiliation(s)
- Sarah Baxendale
- Department of Biomedical Science, Bateson Centre and Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Anzar Asad
- Department of Biomedical Science, Bateson Centre and Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | - Nahal O. Shahidan
- Department of Biomedical Science, Bateson Centre and Neuroscience InstituteUniversity of SheffieldSheffieldUK
| | | | - Tanya T. Whitfield
- Department of Biomedical Science, Bateson Centre and Neuroscience InstituteUniversity of SheffieldSheffieldUK
| |
Collapse
|
10
|
Manini A, Pantoni L. CADASIL from Bench to Bedside: Disease Models and Novel Therapeutic Approaches. Mol Neurobiol 2021; 58:2558-2573. [PMID: 33464533 PMCID: PMC8128844 DOI: 10.1007/s12035-021-02282-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/05/2021] [Indexed: 12/21/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a monogenic disease caused by NOTCH3 mutations and characterized by typical clinical, neuroradiological, and pathological features. NOTCH3 belongs to a family of highly conserved transmembrane receptors rich of epidermal growth factor repeats, mostly expressed in vascular smooth muscle cells and pericytes, which perform essential developmental functions and are involved in tissues maintenance and renewal. To date, no therapeutic option for CADASIL is available except for few symptomatic treatments. Novel in vitro and in vivo models are continuously explored with the aim to investigate underlying pathogenic mechanisms and to test novel therapeutic approaches. In this scenario, knock-out, knock-in, and transgenic mice studies have generated a large amount of information on molecular and biological aspects of CADASIL, despite that they incompletely reproduce the human phenotype. Moreover, the field of in vitro models has been revolutionized in the last two decades by the introduction of induced pluripotent stem cells (iPSCs) technology. As a consequence, novel therapeutic approaches, including immunotherapy, growth factors administration, and antisense oligonucleotides, are currently under investigation. While waiting that further studies confirm the promising results obtained, the data reviewed suggest that our therapeutic approach to the disease could be transformed, generating new hope for the future.
Collapse
Affiliation(s)
- Arianna Manini
- Stroke and Dementia Lab, "Luigi Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Via Giovanni Battista Grassi 74, 20157, Milano, Italy
| | - Leonardo Pantoni
- Stroke and Dementia Lab, "Luigi Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Via Giovanni Battista Grassi 74, 20157, Milano, Italy.
| |
Collapse
|
11
|
Panlilio JM, Aluru N, Hahn ME. Developmental Neurotoxicity of the Harmful Algal Bloom Toxin Domoic Acid: Cellular and Molecular Mechanisms Underlying Altered Behavior in the Zebrafish Model. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:117002. [PMID: 33147070 PMCID: PMC7641300 DOI: 10.1289/ehp6652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
BACKGROUND Harmful algal blooms (HABs) produce potent neurotoxins that threaten human health, but current regulations may not be protective of sensitive populations. Early life exposure to low levels of the HAB toxin domoic acid (DomA) produces long-lasting behavioral deficits in rodent and primate models; however, the mechanisms involved are unknown. The zebrafish is a powerful in vivo vertebrate model system for exploring cellular processes during development and thus may help to elucidate mechanisms of DomA developmental neurotoxicity. OBJECTIVES We used the zebrafish model to investigate how low doses of DomA affect the developing nervous system, including windows of susceptibility to DomA exposure, structural and molecular changes in the nervous system, and the link to behavioral alterations. METHODS To identify potential windows of susceptibility, DomA (0.09-0.18 ng) was delivered to zebrafish through caudal vein microinjection during distinct periods in early neurodevelopment. Following exposure, structural and molecular targets were identified using live imaging of transgenic fish and RNA sequencing. To assess the functional consequences of exposures, we quantified startle behavior in response to acoustic/vibrational stimuli. RESULTS Larvae exposed to DomA at 2 d postfertilization (dpf), but not at 1 or 4 dpf, showed consistent deficits in startle behavior at 7 dpf, including lower responsiveness and altered kinematics. Similarly, myelination in the spinal cord was disorganized after exposure at 2 dpf but not 1 or 4 dpf. Time-lapse imaging revealed disruption of the initial stages of myelination. DomA exposure at 2 dpf down-regulated genes required for maintaining myelin structure and the axonal cytoskeleton. DISCUSSION These results in zebrafish reveal a developmental window of susceptibility to DomA-induced behavioral deficits and identify altered gene expression and disrupted myelin structure as possible mechanisms. The results establish a zebrafish model for investigating the mechanisms of developmental DomA toxicity, including effects with potential relevance to exposed sensitive human populations. https://doi.org/10.1289/EHP6652.
Collapse
Affiliation(s)
- Jennifer M. Panlilio
- Biology Department, Woods Hole Oceanographic Institution (WHOI), Woods Hole, Massachusetts, USA
- Massachusetts Institute of Technology (MIT)–WHOI Joint Graduate Program in Oceanography and Oceanographic Engineering, Department of Earth, Atmospheric and Planetary Sciences, MIT, Cambridge, Massachusetts, USA
- Woods Hole Center for Oceans and Human Health, WHOI, Woods Hole, Massachusetts, USA
| | - Neelakanteswar Aluru
- Biology Department, Woods Hole Oceanographic Institution (WHOI), Woods Hole, Massachusetts, USA
- Woods Hole Center for Oceans and Human Health, WHOI, Woods Hole, Massachusetts, USA
| | - Mark E. Hahn
- Biology Department, Woods Hole Oceanographic Institution (WHOI), Woods Hole, Massachusetts, USA
- Woods Hole Center for Oceans and Human Health, WHOI, Woods Hole, Massachusetts, USA
| |
Collapse
|
12
|
Bradley EC, Cunningham RL, Wilde C, Morgan RK, Klug EA, Letcher SM, Schöneberg T, Monk KR, Liebscher I, Petersen SC. In vivo identification of small molecules mediating Gpr126/Adgrg6 signaling during Schwann cell development. Ann N Y Acad Sci 2019; 1456:44-63. [PMID: 31529518 PMCID: PMC7189964 DOI: 10.1111/nyas.14233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/30/2019] [Accepted: 08/26/2019] [Indexed: 12/13/2022]
Abstract
Gpr126/Adgrg6, an adhesion family G protein-coupled receptor (aGPCR), is required for the development of myelinating Schwann cells in the peripheral nervous system. Myelin supports and insulates vertebrate axons to permit rapid signal propagation throughout the nervous system. In mammals and zebrafish, mutations in Gpr126 arrest Schwann cells at early developmental stages. We exploited the optical and pharmacological tractability of larval zebrafish to uncover drugs that mediate myelination by activating Gpr126 or functioning in parallel. Using a fluorescent marker of mature myelinating glia (Tg[mbp:EGFP-CAAX]), we screened hypomorphic gpr126 mutant larvae for restoration of myelin basic protein (mbp) expression along peripheral nerves following small molecule treatment. Our screens identified five compounds sufficient to promote mbp expression in gpr126 hypomorphs. Using an allelic series of gpr126 mutants, we parsed the ability of small molecules to restore mbp, suggesting differences in drug efficacy dependent on Schwann cell developmental state. Finally, we identify apomorphine hydrochloride as a direct small molecule activator of Gpr126 using combined in vivo/in vitro assays and show that aporphine class compounds promote Schwann cell development in vivo. Our results demonstrate the utility of in vivo screening for aGPCR modulators and identify small molecules that interact with the gpr126-mediated myelination program.
Collapse
Affiliation(s)
| | - Rebecca L. Cunningham
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Caroline Wilde
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Rory K. Morgan
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Emma A. Klug
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
| | | | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Kelly R. Monk
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Sarah C. Petersen
- Department of Neuroscience, Kenyon College, Gambier, OH, USA
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
13
|
Fuentes R, Letelier J, Tajer B, Valdivia LE, Mullins MC. Fishing forward and reverse: Advances in zebrafish phenomics. Mech Dev 2018; 154:296-308. [PMID: 30130581 PMCID: PMC6289646 DOI: 10.1016/j.mod.2018.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/06/2018] [Accepted: 08/17/2018] [Indexed: 12/15/2022]
Abstract
Understanding how the genome instructs the phenotypic characteristics of an organism is one of the major scientific endeavors of our time. Advances in genetics have progressively deciphered the inheritance, identity and biological relevance of genetically encoded information, contributing to the rise of several, complementary omic disciplines. One of them is phenomics, an emergent area of biology dedicated to the systematic multi-scale analysis of phenotypic traits. This discipline provides valuable gene function information to the rapidly evolving field of genetics. Current molecular tools enable genome-wide analyses that link gene sequence to function in multi-cellular organisms, illuminating the genome-phenome relationship. Among vertebrates, zebrafish has emerged as an outstanding model organism for high-throughput phenotyping and modeling of human disorders. Advances in both systematic mutagenesis and phenotypic analyses of embryonic and post-embryonic stages in zebrafish have revealed the function of a valuable collection of genes and the general structure of several complex traits. In this review, we summarize multiple large-scale genetic efforts addressing parental, embryonic, and adult phenotyping in the zebrafish. The genetic and quantitative tools available in the zebrafish model, coupled with the broad spectrum of phenotypes that can be assayed, make it a powerful model for phenomics, well suited for the dissection of genotype-phenotype associations in development, physiology, health and disease.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joaquín Letelier
- Centro Andaluz de Biología del Desarrollo (CSIC/UPO/JA), Seville, Spain; Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Benjamin Tajer
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonardo E Valdivia
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| | - Mary C Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
14
|
Del Giovane A, Ragnini-Wilson A. Targeting Smoothened as a New Frontier in the Functional Recovery of Central Nervous System Demyelinating Pathologies. Int J Mol Sci 2018; 19:E3677. [PMID: 30463396 PMCID: PMC6274747 DOI: 10.3390/ijms19113677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
Abstract
Myelin sheaths on vertebrate axons provide protection, vital support and increase the speed of neuronal signals. Myelin degeneration can be caused by viral, autoimmune or genetic diseases. Remyelination is a natural process that restores the myelin sheath and, consequently, neuronal function after a demyelination event, preventing neurodegeneration and thereby neuron functional loss. Pharmacological approaches to remyelination represent a promising new frontier in the therapy of human demyelination pathologies and might provide novel tools to improve adaptive myelination in aged individuals. Recent phenotypical screens have identified agonists of the atypical G protein-coupled receptor Smoothened and inhibitors of the glioma-associated oncogene 1 as being amongst the most potent stimulators of oligodendrocyte precursor cell (OPC) differentiation in vitro and remyelination in the central nervous system (CNS) of mice. Here, we discuss the current state-of-the-art of studies on the role of Sonic Hedgehog reactivation during remyelination, referring readers to other reviews for the role of Hedgehog signaling in cancer and stem cell maintenance.
Collapse
Affiliation(s)
- Alice Del Giovane
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| | - Antonella Ragnini-Wilson
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| |
Collapse
|
15
|
Cunningham RL, Herbert AL, Harty BL, Ackerman SD, Monk KR. Mutations in dock1 disrupt early Schwann cell development. Neural Dev 2018; 13:17. [PMID: 30089513 PMCID: PMC6083577 DOI: 10.1186/s13064-018-0114-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/20/2018] [Indexed: 01/29/2023] Open
Abstract
Background In the peripheral nervous system (PNS), specialized glial cells called Schwann cells produce myelin, a lipid-rich insulating sheath that surrounds axons and promotes rapid action potential propagation. During development, Schwann cells must undergo extensive cytoskeletal rearrangements in order to become mature, myelinating Schwann cells. The intracellular mechanisms that drive Schwann cell development, myelination, and accompanying cell shape changes are poorly understood. Methods Through a forward genetic screen in zebrafish, we identified a mutation in the atypical guanine nucleotide exchange factor, dock1, that results in decreased myelination of peripheral axons. Rescue experiments and complementation tests with newly engineered alleles confirmed that mutations in dock1 cause defects in myelination of the PNS. Whole mount in situ hybridization, transmission electron microscopy, and live imaging were used to fully define mutant phenotypes. Results We show that Schwann cells in dock1 mutants can appropriately migrate and are not decreased in number, but exhibit delayed radial sorting and decreased myelination during early stages of development. Conclusions Together, our results demonstrate that mutations in dock1 result in defects in Schwann cell development and myelination. Specifically, loss of dock1 delays radial sorting and myelination of peripheral axons in zebrafish. Electronic supplementary material The online version of this article (10.1186/s13064-018-0114-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rebecca L Cunningham
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Amy L Herbert
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Breanne L Harty
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Vollum Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Sarah D Ackerman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Institute of Neuroscience, University of Oregon, Eugene, OR, 97403, USA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Vollum Institute, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
16
|
Earley AM, Dixon CT, Shiau CE. Genetic analysis of zebrafish homologs of human FOXQ1, foxq1a and foxq1b, in innate immune cell development and bacterial host response. PLoS One 2018; 13:e0194207. [PMID: 29534099 PMCID: PMC5849333 DOI: 10.1371/journal.pone.0194207] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/27/2018] [Indexed: 01/01/2023] Open
Abstract
FOXQ1 is a member of the forkhead-box transcription factor family that has important functions in development, cancer, aging, and many cellular processes. The role of FOXQ1 in cancer biology has raised intense interest, yet much remains poorly understood. We investigated the possible function of the two zebrafish orthologs (foxq1a and foxq1b) of human FOXQ1 in innate immune cell development and function. We employed CRISPR-Cas9 targeted mutagenesis to create null mutations of foxq1a and foxq1b in zebrafish. Using a combination of molecular, cellular, and embryological approaches, we characterized single and double foxq1a bcz11 and foxq1b bcz18 mutants. This study provides the first genetic mutant analyses of zebrafish foxq1a and foxq1b. Interestingly, we found that foxq1a, but not foxq1b, was transcriptionally regulated during a bacterial response, while the expression of foxq1a was detected in sorted macrophages and upregulated in foxq1a-deficient mutants. However, the transcriptional response to E. coli challenge of foxq1a and foxq1b mutants was not significantly different from that of their wildtype control siblings. Our data shows that foxq1a may have a role in modulating bacterial response, while both foxq1a and foxq1b are not required for the development of macrophages, neutrophils, and microglia. Considering the implicated role of FOXQ1 in a vast number of cancers and biological processes, the foxq1a and foxq1b null mutants from this study provide useful genetic models to further investigate FOXQ1 functions.
Collapse
Affiliation(s)
- Alison M. Earley
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Cameron T. Dixon
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Celia E. Shiau
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
17
|
Abstract
In situ hybridization enables visualization of mRNA localization, and immunohistochemistry enables visualization of protein localization within a tissue or organism. Both techniques have been extensively utilized in zebrafish (Thisse et al., Development 119:1203-1215, 1993; Dutton et al., Development 128:4113-4125, 2001; Gilmour et al., Neuron 34:577-588, 2002; Lyons et al., Curr Biol 15:513-524, 2005) including for visualization of mRNA localization in Schwann cells (Lyons et al., Curr Biol 15:513-524, 2005; Monk et al., Science 325:1402-1405, 2009). For in situ hybridization, here, we outline how to generate RNA probes, conduct whole mount in situ hybridization for larvae, and list RNA probes that label different stages of Schwann cell development in zebrafish. For immunohistochemistry, the protocol we outline can be used to mark Schwann cells of sensory and motor nerves to examine properties such as developmental stage, morphology, proliferation, and apoptosis.
Collapse
|
18
|
Harty BL, Monk KR. Unwrapping the unappreciated: recent progress in Remak Schwann cell biology. Curr Opin Neurobiol 2017; 47:131-137. [PMID: 29096241 DOI: 10.1016/j.conb.2017.10.003] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/11/2017] [Accepted: 10/06/2017] [Indexed: 11/19/2022]
Abstract
Schwann cells (SCs) are specialized glial cells that myelinate and protect axons in the peripheral nervous system (PNS). Although myelinating SCs are more commonly studied, the PNS also contains a variety of non-myelinating SCs, including but not limited to Remak SCs (RSCs), terminal SCs, enteric glia. Although the field currently lacks many robust tools for interrogating the functions of non-myelinating SCs, recent evidence suggests that, like their myelinating counterparts, non-myelinating SCs are critical for proper PNS function. In this review, we focus specifically on RSCs and highlight recent advances in understanding regulators of RSC development, function, and participation in PNS regeneration.
Collapse
Affiliation(s)
- Breanne L Harty
- Department of Developmental Biology, Washington University School of Medicine, 660 S Euclid Ave, St Louis, MO 63110, USA.
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, 660 S Euclid Ave, St Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, USA.
| |
Collapse
|
19
|
Abstract
Fell et al. deleted KIF1Bβ in the mouse sympathetic nervous system and observed impaired sympathetic nervous function and misexpression of genes required for sympathoadrenal lineage differentiation. They discovered that KIF1Bβ is required for NGF-dependent neuronal differentiation through anterograde transport of the NGF receptor TRKA. We recently identified pathogenic KIF1Bβ mutations in sympathetic nervous system malignancies that are defective in developmental apoptosis. Here we deleted KIF1Bβ in the mouse sympathetic nervous system and observed impaired sympathetic nervous function and misexpression of genes required for sympathoadrenal lineage differentiation. We discovered that KIF1Bβ is required for nerve growth factor (NGF)-dependent neuronal differentiation through anterograde transport of the NGF receptor TRKA. Moreover, pathogenic KIF1Bβ mutations identified in neuroblastoma impair TRKA transport. Expression of neuronal differentiation markers is ablated in both KIF1Bβ-deficient mouse neuroblasts and human neuroblastomas that lack KIF1Bβ. Transcriptomic analyses show that unfavorable neuroblastomas resemble mouse sympathetic neuroblasts lacking KIF1Bβ independent of MYCN amplification and the loss of genes neighboring KIF1B on chromosome 1p36. Thus, defective precursor cell differentiation, a common trait of aggressive childhood malignancies, is a pathogenic effect of KIF1Bβ loss in neuroblastomas. Furthermore, neuropathy-associated KIF1Bβ mutations impede cargo transport, providing a direct link between neuroblastomas and neurodegeneration.
Collapse
|
20
|
Whole Genome Sequencing-Based Mapping and Candidate Identification of Mutations from Fixed Zebrafish Tissue. G3-GENES GENOMES GENETICS 2017; 7:3415-3425. [PMID: 28855284 PMCID: PMC5633390 DOI: 10.1534/g3.117.300212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
As forward genetic screens in zebrafish become more common, the number of mutants that cannot be identified by gross morphology or through transgenic approaches, such as many nervous system defects, has also increased. Screening for these difficult-to-visualize phenotypes demands techniques such as whole-mount in situ hybridization (WISH) or antibody staining, which require tissue fixation. To date, fixed tissue has not been amenable for generating libraries for whole genome sequencing (WGS). Here, we describe a method for using genomic DNA from fixed tissue and a bioinformatics suite for WGS-based mapping of zebrafish mutants. We tested our protocol using two known zebrafish mutant alleles, gpr126st49 and egr2bfh227, both of which cause myelin defects. As further proof of concept we mapped a novel mutation, stl64, identified in a zebrafish WISH screen for myelination defects. We linked stl64 to chromosome 1 and identified a candidate nonsense mutation in the F-box and WD repeat domain containing 7 (fbxw7) gene. Importantly, stl64 mutants phenocopy previously described fbxw7vu56 mutants, and knockdown of fbxw7 in wild-type animals produced similar defects, demonstrating that stl64 disrupts fbxw7. Together, these data show that our mapping protocol can map and identify causative lesions in mutant screens that require tissue fixation for phenotypic analysis.
Collapse
|
21
|
Cole KLH, Early JJ, Lyons DA. Drug discovery for remyelination and treatment of MS. Glia 2017; 65:1565-1589. [PMID: 28618073 DOI: 10.1002/glia.23166] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 12/19/2022]
Abstract
Glia constitute the majority of the cells in our nervous system, yet there are currently no drugs that target glia for the treatment of disease. Given ongoing discoveries of the many roles of glia in numerous diseases of the nervous system, this is likely to change in years to come. Here we focus on the possibility that targeting the oligodendrocyte lineage to promote regeneration of myelin (remyelination) represents a therapeutic strategy for the treatment of the demyelinating disease multiple sclerosis, MS. We discuss how hypothesis driven studies have identified multiple targets and pathways that can be manipulated to promote remyelination in vivo, and how this work has led to the first ever remyelination clinical trials. We also highlight how recent chemical discovery screens have identified a host of small molecule compounds that promote oligodendrocyte differentiation in vitro. Some of these compounds have also been shown to promote myelin regeneration in vivo, with one already being trialled in humans. Promoting oligodendrocyte differentiation and remyelination represents just one potential strategy for the treatment of MS. The pathology of MS is complex, and its complete amelioration may require targeting multiple biological processes in parallel. Therefore, we present an overview of new technologies and models for phenotypic analyses and screening that can be exploited to study complex cell-cell interactions in in vitro and in vivo systems. Such technological platforms will provide insight into fundamental mechanisms and increase capacities for drug-discovery of relevance to glia and currently intractable disorders of the CNS.
Collapse
Affiliation(s)
- Katy L H Cole
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - Jason J Early
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| | - David A Lyons
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, EH16 4SB, United Kingdom
| |
Collapse
|
22
|
Abstract
Myelination by oligodendrocytes in the central nervous system (CNS) and Schwann cells in the peripheral nervous system is essential for nervous system function and health. Despite its importance, we have a relatively poor understanding of the molecular and cellular mechanisms that regulate myelination in the living animal, particularly in the CNS. This is partly due to the fact that myelination commences around birth in mammals, by which time the CNS is complex and largely inaccessible, and thus very difficult to image live in its intact form. As a consequence, in recent years much effort has been invested in the use of smaller, simpler, transparent model organisms to investigate mechanisms of myelination in vivo. Although the majority of such studies have employed zebrafish, the Xenopus tadpole also represents an important complementary system with advantages for investigating myelin biology in vivo. Here we review how the natural features of zebrafish embryos and larvae and Xenopus tadpoles make them ideal systems for experimentally interrogating myelination by live imaging. We outline common transgenic technologies used to generate zebrafish and Xenopus that express fluorescent reporters, which can be used to image myelination. We also provide an extensive overview of the imaging modalities most commonly employed to date to image the nervous system in these transparent systems, and also emerging technologies that we anticipate will become widely used in studies of zebrafish and Xenopus myelination in the near future.
Collapse
Affiliation(s)
- Jenea M Bin
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - David A Lyons
- Centre for Neuroregeneration, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
23
|
Fontenas L, De Santis F, Di Donato V, Degerny C, Chambraud B, Del Bene F, Tawk M. Neuronal Ndrg4 Is Essential for Nodes of Ranvier Organization in Zebrafish. PLoS Genet 2016; 12:e1006459. [PMID: 27902705 PMCID: PMC5130175 DOI: 10.1371/journal.pgen.1006459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 11/03/2016] [Indexed: 11/19/2022] Open
Abstract
Axon ensheathment by specialized glial cells is an important process for fast propagation of action potentials. The rapid electrical conduction along myelinated axons is mainly due to its saltatory nature characterized by the accumulation of ion channels at the nodes of Ranvier. However, how these ion channels are transported and anchored along axons is not fully understood. We have identified N-myc downstream-regulated gene 4, ndrg4, as a novel factor that regulates sodium channel clustering in zebrafish. Analysis of chimeric larvae indicates that ndrg4 functions autonomously within neurons for sodium channel clustering at the nodes. Molecular analysis of ndrg4 mutants shows that expression of snap25 and nsf are sharply decreased, revealing a role of ndrg4 in controlling vesicle exocytosis. This uncovers a previously unknown function of ndrg4 in regulating vesicle docking and nodes of Ranvier organization, at least through its ability to finely tune the expression of the t-SNARE/NSF machinery. Myelination is an important process that enables fast propagation of action potential along the axons. Schwann cells (SCs) are the specialized glial cells that ensure the ensheathment of the corresponding axons in the Peripheral Nervous System. In order to do so, SCs and axons need to communicate to organize the myelinating segments and the clustering of sodium channels at the nodes of Ranvier. We have investigated the early events of myelination in the zebrafish embryo. We here identify ndrg4 as a novel neuronal factor essential for sodium channel clustering at the nodes. Immuno-labeling analysis show defective vesicle patterning along the axons of ndrg4 mutants, while timelapse experiments monitoring the presence and the transport of these vesicles reveal a normal behavior. Molecular analysis unravels a novel function of ndrg4 in controlling the expression of the t-SNARE/NSF machinery required for vesicle docking and release. However, inhibiting specifically regulated synaptic vesicle release does not lead to sodium channel clustering defects. We thus propose that ndrg4 can regulate this process, at least partially, through its ability to regulate the expression of key components of the t-SNARE/NSF machinery, responsible for clustering of sodium channels along myelinated axons.
Collapse
Affiliation(s)
- Laura Fontenas
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
| | | | | | - Cindy Degerny
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
| | - Béatrice Chambraud
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
| | | | - Marcel Tawk
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
- * E-mail:
| |
Collapse
|
24
|
Abstract
Myelin is a lipid-rich sheath formed by the spiral wrapping of specialized glial cells around axon segments. Myelinating glia allow for rapid transmission of nerve impulses and metabolic support of axons, and the absence of or disruption to myelin results in debilitating motor, cognitive, and emotional deficits in humans. Because myelin is a jawed vertebrate innovation, zebrafish are one of the simplest vertebrate model systems to study the genetics and development of myelinating glia. The morphogenetic cellular movements and genetic program that drive myelination are conserved between zebrafish and mammals, and myelin develops rapidly in zebrafish larvae, within 3-5days postfertilization. Myelin ultrastructure can be visualized in the zebrafish from larval to adult stages via transmission electron microscopy, and the dynamic development of myelinating glial cells may be observed in vivo via transgenic reporter lines in zebrafish larvae. Zebrafish are amenable to genetic and pharmacological screens, and screens for myelinating glial phenotypes have revealed both genes and drugs that promote myelin development, many of which are conserved in mammalian glia. Recently, zebrafish have been employed as a model to understand the complex dynamics of myelinating glia during development and regeneration. In this chapter, we describe these key methodologies and recent insights into mechanisms that regulate myelination using the zebrafish model.
Collapse
Affiliation(s)
- M D'Rozario
- Washington University School of Medicine, St. Louis, MO, United States
| | - K R Monk
- Washington University School of Medicine, St. Louis, MO, United States; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | | |
Collapse
|
25
|
Mogha A, D'Rozario M, Monk KR. G Protein-Coupled Receptors in Myelinating Glia. Trends Pharmacol Sci 2016; 37:977-987. [PMID: 27670389 DOI: 10.1016/j.tips.2016.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022]
Abstract
The G protein-coupled receptor (GPCR) superfamily represents the largest class of functionally selective drug targets for disease modulation and therapy. GPCRs have been studied in great detail in central nervous system (CNS) neurons, but these important molecules have been relatively understudied in glia. In recent years, however, exciting new roles for GPCRs in glial cell biology have emerged. We focus here on the key roles of GPCRs in a specialized subset of glia, myelinating glia. We highlight recent work firmly establishing GPCRs as regulators of myelinating glial cell development and myelin repair. These advances expand our understanding of myelinating glial cell biology and underscore the utility of targeting GPCRs to promote myelin repair in human disease.
Collapse
Affiliation(s)
- Amit Mogha
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mitchell D'Rozario
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Zada D, Tovin A, Lerer-Goldshtein T, Appelbaum L. Pharmacological treatment and BBB-targeted genetic therapy for MCT8-dependent hypomyelination in zebrafish. Dis Model Mech 2016; 9:1339-1348. [PMID: 27664134 PMCID: PMC5117236 DOI: 10.1242/dmm.027227] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/19/2016] [Indexed: 12/31/2022] Open
Abstract
Hypomyelination is a key symptom of Allan-Herndon-Dudley syndrome (AHDS), a psychomotor retardation associated with mutations in the thyroid-hormone (TH) transporter MCT8 (monocarboxylate transporter 8). AHDS is characterized by severe intellectual deficiency, neuromuscular impairment and brain hypothyroidism. In order to understand the mechanism for TH-dependent hypomyelination, we developed an mct8 mutant (mct8-/-) zebrafish model. The quantification of genetic markers for oligodendrocyte progenitor cells (OPCs) and mature oligodendrocytes revealed reduced differentiation of OPCs into oligodendrocytes in mct8-/- larvae and adults. Live imaging of single glial cells showed that the number of oligodendrocytes and the length of their extensions are reduced, and the number of peripheral Schwann cells is increased, in mct8-/- larvae compared with wild type. Pharmacological analysis showed that TH analogs and clemastine partially rescued the hypomyelination in the CNS of mct8-/- larvae. Intriguingly, triiodothyronine (T3) treatment rescued hypomyelination in mct8-/- embryos before the maturation of the blood-brain barrier (BBB), but did not affect hypomyelination in older larvae. Thus, we expressed Mct8-tagRFP in the endothelial cells of the vascular system and showed that even relatively weak mosaic expression completely rescued hypomyelination in mct8-/- larvae. These results suggest potential pharmacological treatments and BBB-targeted gene therapy that can enhance myelination in AHDS and possibly in other TH-dependent brain disorders.
Collapse
Affiliation(s)
- David Zada
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Adi Tovin
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Tali Lerer-Goldshtein
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|
27
|
Küffer A, Lakkaraju AKK, Mogha A, Petersen SC, Airich K, Doucerain C, Marpakwar R, Bakirci P, Senatore A, Monnard A, Schiavi C, Nuvolone M, Grosshans B, Hornemann S, Bassilana F, Monk KR, Aguzzi A. The prion protein is an agonistic ligand of the G protein-coupled receptor Adgrg6. Nature 2016; 536:464-8. [PMID: 27501152 PMCID: PMC5499706 DOI: 10.1038/nature19312] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/19/2016] [Indexed: 12/23/2022]
Abstract
Ablation of the cellular prion protein PrP(C) leads to a chronic demyelinating polyneuropathy affecting Schwann cells. Neuron-restricted expression of PrP(C) prevents the disease, suggesting that PrP(C) acts in trans through an unidentified Schwann cell receptor. Here we show that the cAMP concentration in sciatic nerves from PrP(C)-deficient mice is reduced, suggesting that PrP(C) acts via a G protein-coupled receptor (GPCR). The amino-terminal flexible tail (residues 23-120) of PrP(C) triggered a concentration-dependent increase in cAMP in primary Schwann cells, in the Schwann cell line SW10, and in HEK293T cells overexpressing the GPCR Adgrg6 (also known as Gpr126). By contrast, naive HEK293T cells and HEK293T cells expressing several other GPCRs did not react to the flexible tail, and ablation of Gpr126 from SW10 cells abolished the flexible tail-induced cAMP response. The flexible tail contains a polycationic cluster (KKRPKPG) similar to the GPRGKPG motif of the Gpr126 agonist type-IV collagen. A KKRPKPG-containing PrPC-derived peptide (FT(23-50)) sufficed to induce a Gpr126-dependent cAMP response in cells and mice, and improved myelination in hypomorphic gpr126 mutant zebrafish (Danio rerio). Substitution of the cationic residues with alanines abolished the biological activity of both FT(23-50) and the equivalent type-IV collagen peptide. We conclude that PrP(C) promotes myelin homeostasis through flexible tail-mediated Gpr126 agonism. As well as clarifying the physiological role of PrP(C), these observations are relevant to the pathogenesis of demyelinating polyneuropathies--common debilitating diseases for which there are limited therapeutic options.
Collapse
Affiliation(s)
- Alexander Küffer
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Asvin KK Lakkaraju
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Amit Mogha
- Washington University School of Medicine, Department of Developmental Biology and Hope Center for Neurological Disorders, 660 South Euclid Avenue, Campus Box 8103, St. Louis, MO 63110
| | - Sarah C. Petersen
- Washington University School of Medicine, Department of Developmental Biology and Hope Center for Neurological Disorders, 660 South Euclid Avenue, Campus Box 8103, St. Louis, MO 63110
| | - Kristina Airich
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Cédric Doucerain
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | | | - Pamela Bakirci
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Assunta Senatore
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Arnaud Monnard
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Carmen Schiavi
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Mario Nuvolone
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | - Bianka Grosshans
- Novartis Institutes of Biomedical Research, CH-4056 Basel, Switzerland
| | - Simone Hornemann
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
| | | | - Kelly R. Monk
- Washington University School of Medicine, Department of Developmental Biology and Hope Center for Neurological Disorders, 660 South Euclid Avenue, Campus Box 8103, St. Louis, MO 63110
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, CH-8091 Zürich, Switzerland
- Corresponding author: Adriano Aguzzi, Institute of Neuropathology, University of Zurich, Schmelzbergstrasse 12, CH-8091 Zurich, Switzerland, Tel: +41-44-255-2107,
| |
Collapse
|
28
|
Abstract
In the nervous system, axons transmit information in the form of electrical impulses over long distances. The speed of impulse conduction is enhanced by myelin, a lipid-rich membrane that wraps around axons. Myelin also is required for the long-term health of axons by providing metabolic support. Accordingly, myelin deficiencies are implicated in a wide range of neurodevelopmental and neuropsychiatric disorders, intellectual disabilities, and neurodegenerative conditions. Central nervous system myelin is formed by glial cells called oligodendrocytes. During development, oligodendrocyte precursor cells migrate from their origins to their target axons, extend long membrane processes that wrap axons, and produce the proteins and lipids that provide myelin membrane with its unique characteristics. Myelination is a dynamic process that involves intricate interactions between multiple cell types. Therefore, an in vivo myelination model, such as the zebrafish, which allows for live observation of cell dynamics and cell-to-cell interactions, is well suited for investigating oligodendrocyte development. Zebrafish offer several advantages to investigating myelination, including the use of transgenic reporter lines, live imaging, forward genetic screens, chemical screens, and reverse genetic approaches. This chapter will describe how these tools and approaches have provided new insights into the regulatory mechanisms that guide myelination.
Collapse
Affiliation(s)
- E S Mathews
- University of Colorado School of Medicine, Aurora, CO, United States
| | - B Appel
- University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
29
|
Möbius W, Nave KA, Werner HB. Electron microscopy of myelin: Structure preservation by high-pressure freezing. Brain Res 2016; 1641:92-100. [PMID: 26920467 DOI: 10.1016/j.brainres.2016.02.027] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/16/2016] [Indexed: 10/24/2022]
Abstract
Electron microscopic visualization of nervous tissue morphology is crucial when aiming to understand the biogenesis and structure of myelin in healthy and pathological conditions. However, accurate interpretation of electron micrographs requires excellent tissue preservation. In this short review we discuss the recent utilization of tissue fixation by high-pressure freezing and freeze-substitution, which now supplements aldehyde fixation in the preparation of samples for electron microscopy of myelin. Cryofixation has proven well suited to yield both, improved contrast and excellent preservation of structural detail of the axon/myelin-unit in healthy and mutant mice and can also be applied to other model organisms, including aquatic species. This article is part of a Special Issue entitled SI: Myelin Evolution.
Collapse
Affiliation(s)
- Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany; Center for Nanoscale Microscopy and Molecular Physiology of the Brain, 37075 Göttingen, Germany.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany.
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
30
|
Shen K, Sidik H, Talbot WS. The Rag-Ragulator Complex Regulates Lysosome Function and Phagocytic Flux in Microglia. Cell Rep 2016; 14:547-559. [PMID: 26774477 PMCID: PMC4731305 DOI: 10.1016/j.celrep.2015.12.055] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/24/2015] [Accepted: 12/09/2015] [Indexed: 01/07/2023] Open
Abstract
Microglia are resident macrophages of the CNS that are essential for phagocytosis of apoptotic neurons and weak synapses during development. We show that RagA and Lamtor4, two components of the Rag-Ragulator complex, are essential regulators of lysosomes in microglia. In zebrafish lacking RagA function, microglia exhibit an expanded lysosomal compartment, but they are unable to properly digest apoptotic neuronal debris. Previous biochemical studies have placed the Rag-Ragulator complex upstream of mTORC1 activation in response to cellular nutrient availability. Nonetheless, RagA and mTOR mutant zebrafish have distinct phenotypes, indicating that the Rag-Ragulator complex has functions independent of mTOR signaling. Our analysis reveals an essential role of the Rag-Ragulator complex in proper lysosome function and phagocytic flux in microglia.
Collapse
Affiliation(s)
- Kimberle Shen
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Harwin Sidik
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William S Talbot
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
31
|
The scales and tales of myelination: using zebrafish and mouse to study myelinating glia. Brain Res 2015; 1641:79-91. [PMID: 26498880 DOI: 10.1016/j.brainres.2015.10.011] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 01/06/2023]
Abstract
Myelin, the lipid-rich sheath that insulates axons to facilitate rapid conduction of action potentials, is an evolutionary innovation of the jawed-vertebrate lineage. Research efforts aimed at understanding the molecular mechanisms governing myelination have primarily focused on rodent models; however, with the advent of the zebrafish model system in the late twentieth century, the use of this genetically tractable, yet simpler vertebrate for studying myelination has steadily increased. In this review, we compare myelinating glial cell biology during development and regeneration in zebrafish and mouse and enumerate the advantages and disadvantages of using each model to study myelination. This article is part of a Special Issue entitled SI: Myelin Evolution.
Collapse
|
32
|
Sidik H, Talbot WS. A zinc finger protein that regulates oligodendrocyte specification, migration and myelination in zebrafish. Development 2015; 142:4119-28. [PMID: 26459222 DOI: 10.1242/dev.128215] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/01/2015] [Indexed: 12/21/2022]
Abstract
Precise control of oligodendrocyte migration and development is crucial for myelination of axons in the central nervous system (CNS), but important questions remain unanswered about the mechanisms controlling these processes. In a zebrafish screen for myelination mutants, we identified a mutation in zinc finger protein 16-like (znf16l). znf16l mutant larvae have reduced myelin basic protein (mbp) expression and reduced CNS myelin. Marker, time-lapse and ultrastructural studies indicated that oligodendrocyte specification, migration and myelination are disrupted in znf16l mutants. Transgenic studies indicated that znf16l acts autonomously in oligodendrocytes. Expression of Zfp488 from mouse rescued mbp expression in znf16l mutants, indicating that these homologs have overlapping functions. Our results defined the function of a new zinc finger protein with specific function in oligodendrocyte specification, migration and myelination in the developing CNS.
Collapse
Affiliation(s)
- Harwin Sidik
- Department of Developmental Biology, Stanford University, CA 94305, USA
| | - William S Talbot
- Department of Developmental Biology, Stanford University, CA 94305, USA
| |
Collapse
|
33
|
Czopka T. Insights into mechanisms of central nervous system myelination using zebrafish. Glia 2015; 64:333-49. [PMID: 26250418 DOI: 10.1002/glia.22897] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/14/2015] [Accepted: 07/15/2015] [Indexed: 12/12/2022]
Abstract
Myelin is the multi-layered membrane that surrounds most axons and is produced by oligodendrocytes in the central nervous system (CNS). In addition to its important role in enabling rapid nerve conduction, it has become clear in recent years that myelin plays additional vital roles in CNS function. Myelinating oligodendrocytes provide metabolic support to axons and active myelination is even involved in regulating forms of learning and memory formation. However, there are still large gaps in our understanding of how myelination by oligodendrocytes is regulated. The small tropical zebrafish has become an increasingly popular model organism to investigate many aspects of nervous system formation, function, and regeneration. This is mainly due to two approaches for which the zebrafish is an ideally suited vertebrate model--(1) in vivo live cell imaging using vital dyes and genetically encoded reporters, and (2) gene and target discovery using unbiased screens. This review summarizes how the use of zebrafish has helped understand mechanisms of oligodendrocyte behavior and myelination in vivo and discusses the potential use of zebrafish to shed light on important future questions relating to myelination in the context of CNS development, function and repair.
Collapse
Affiliation(s)
- Tim Czopka
- Institute of Neuronal Cell Biology, Technische Universität München, Munich, Germany
| |
Collapse
|
34
|
Abstract
Myelination of axons in the nervous system of vertebrates enables fast, saltatory impulse propagation, one of the best-understood concepts in neurophysiology. However, it took a long while to recognize the mechanistic complexity both of myelination by oligodendrocytes and Schwann cells and of their cellular interactions. In this review, we highlight recent advances in our understanding of myelin biogenesis, its lifelong plasticity, and the reciprocal interactions of myelinating glia with the axons they ensheath. In the central nervous system, myelination is also stimulated by axonal activity and astrocytes, whereas myelin clearance involves microglia/macrophages. Once myelinated, the long-term integrity of axons depends on glial supply of metabolites and neurotrophic factors. The relevance of this axoglial symbiosis is illustrated in normal brain aging and human myelin diseases, which can be studied in corresponding mouse models. Thus, myelinating cells serve a key role in preserving the connectivity and functions of a healthy nervous system.
Collapse
Affiliation(s)
- Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, D-37075 Göttingen, Germany; ,
| | | |
Collapse
|
35
|
Petersen SC, Luo R, Liebscher I, Giera S, Jeong SJ, Mogha A, Ghidinelli M, Feltri ML, Schöneberg T, Piao X, Monk KR. The adhesion GPCR GPR126 has distinct, domain-dependent functions in Schwann cell development mediated by interaction with laminin-211. Neuron 2015; 85:755-69. [PMID: 25695270 DOI: 10.1016/j.neuron.2014.12.057] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 11/12/2014] [Accepted: 12/17/2014] [Indexed: 10/24/2022]
Abstract
Myelin ensheathes axons to allow rapid propagation of action potentials and proper nervous system function. In the peripheral nervous system, Schwann cells (SCs) radially sort axons into a 1:1 relationship before wrapping an axonal segment to form myelin. SC myelination requires the adhesion G protein-coupled receptor GPR126, which undergoes autoproteolytic cleavage into an N-terminal fragment (NTF) and a seven-transmembrane-containing C-terminal fragment (CTF). Here we show that GPR126 has domain-specific functions in SC development whereby the NTF is necessary and sufficient for axon sorting, whereas the CTF promotes wrapping through cAMP elevation. These biphasic roles of GPR126 are governed by interactions with Laminin-211, which we define as a novel ligand for GPR126 that modulates receptor signaling via a tethered agonist. Our work suggests a model in which Laminin-211 mediates GPR126-induced cAMP levels to control early and late stages of SC development.
Collapse
Affiliation(s)
- Sarah C Petersen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rong Luo
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ines Liebscher
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Stefanie Giera
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Sung-Jin Jeong
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amit Mogha
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Monica Ghidinelli
- Department of Biochemistry, University of Buffalo, The State University of New York, Buffalo, NY 14023, USA
| | - M Laura Feltri
- Department of Biochemistry, University of Buffalo, The State University of New York, Buffalo, NY 14023, USA
| | - Torsten Schöneberg
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Xianhua Piao
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
36
|
Xiao Y, Faucherre A, Pola-Morell L, Heddleston JM, Liu TL, Chew TL, Sato F, Sehara-Fujisawa A, Kawakami K, López-Schier H. High-resolution live imaging reveals axon-glia interactions during peripheral nerve injury and repair in zebrafish. Dis Model Mech 2015; 8:553-64. [PMID: 26035865 PMCID: PMC4457030 DOI: 10.1242/dmm.018184] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 03/24/2015] [Indexed: 12/22/2022] Open
Abstract
Neural damage is a devastating outcome of physical trauma. The glia are one of the main effectors of neuronal repair in the nervous system, but the dynamic interactions between peripheral neurons and Schwann cells during injury and regeneration remain incompletely characterized. Here, we combine laser microsurgery, genetic analysis, high-resolution intravital imaging and lattice light-sheet microscopy to study the interaction between Schwann cells and sensory neurons in a zebrafish model of neurotrauma. We found that chronic denervation by neuronal ablation leads to Schwann-cell death, whereas acute denervation by axonal severing does not affect the overall complexity and architecture of the glia. Neuronal-circuit regeneration begins when Schwann cells extend bridging processes to close the injury gap. Regenerating axons grow faster and directionally after the physiological clearing of distal debris by the Schwann cells. This might facilitate circuit repair by ensuring that axons are guided through unoccupied spaces within bands of Büngner towards their original peripheral target. Accordingly, in the absence of Schwann cells, regenerating axons are misrouted, impairing the re-innervation of sensory organs. Our results indicate that regenerating axons use haptotaxis as a directional cue during the reconstitution of a neural circuit. These findings have implications for therapies aimed at neurorepair, which will benefit from preserving the architecture of the peripheral glia during periods of denervation. Summary: Schwann cells are important components of the peripheral glia. We use microsurgery and high-resolution live imaging to show how Schwann cells control the regeneration of a sensorineural circuit.
Collapse
Affiliation(s)
- Yan Xiao
- Research Unit Sensory Biology & Organogenesis, Helmholtz Zentrum München, 85764 Munich, Germany
| | - Adèle Faucherre
- Cell & Developmental Biology, Centre for Genomic Regulation, 08003 Barcelona, Spain
| | - Laura Pola-Morell
- Cell & Developmental Biology, Centre for Genomic Regulation, 08003 Barcelona, Spain
| | - John M Heddleston
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Tsung-Li Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Teng-Leong Chew
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Fuminori Sato
- Institute for Frontier Medical Sciences, Kyoto University, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Atsuko Sehara-Fujisawa
- Institute for Frontier Medical Sciences, Kyoto University, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, The Graduate University for Advanced Studies (Sokendai), 1111 Yata, Mishima, Shizuoka 411-8540, Japan Department of Genetics, The Graduate University for Advanced Studies (Sokendai), 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| | - Hernán López-Schier
- Research Unit Sensory Biology & Organogenesis, Helmholtz Zentrum München, 85764 Munich, Germany Cell & Developmental Biology, Centre for Genomic Regulation, 08003 Barcelona, Spain
| |
Collapse
|
37
|
Liebscher I, Schön J, Petersen SC, Fischer L, Auerbach N, Demberg LM, Mogha A, Cöster M, Simon KU, Rothemund S, Monk KR, Schöneberg T. A tethered agonist within the ectodomain activates the adhesion G protein-coupled receptors GPR126 and GPR133. Cell Rep 2014; 9:2018-26. [PMID: 25533341 DOI: 10.1016/j.celrep.2014.11.036] [Citation(s) in RCA: 209] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/10/2014] [Accepted: 11/22/2014] [Indexed: 01/08/2023] Open
Abstract
Adhesion G protein-coupled receptors (aGPCRs) comprise the second largest yet least studied class of the GPCR superfamily. aGPCRs are involved in many developmental processes and immune and synaptic functions, but the mode of their signal transduction is unclear. Here, we show that a short peptide sequence (termed the Stachel sequence) within the ectodomain of two aGPCRs (GPR126 and GPR133) functions as a tethered agonist. Upon structural changes within the receptor ectodomain, this intramolecular agonist is exposed to the seven-transmembrane helix domain, which triggers G protein activation. Our studies show high specificity of a given Stachel sequence for its receptor. Finally, the function of Gpr126 is abrogated in zebrafish with a mutated Stachel sequence, and signaling is restored in hypomorphic gpr126 zebrafish mutants upon exogenous Stachel peptide application. These findings illuminate a mode of aGPCR activation and may prompt the development of specific ligands for this currently untargeted GPCR family.
Collapse
Affiliation(s)
- Ines Liebscher
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; Novo Nordisk Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Julia Schön
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Sarah C Petersen
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Liane Fischer
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Nina Auerbach
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lilian Marie Demberg
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Amit Mogha
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maxi Cöster
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Kay-Uwe Simon
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Sven Rothemund
- Core Unit Peptide Technologies, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Torsten Schöneberg
- Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
38
|
Liebscher I, Ackley B, Araç D, Ariestanti DM, Aust G, Bae BI, Bista BR, Bridges JP, Duman JG, Engel FB, Giera S, Goffinet AM, Hall RA, Hamann J, Hartmann N, Lin HH, Liu M, Luo R, Mogha A, Monk KR, Peeters MC, Prömel S, Ressl S, Schiöth HB, Sigoillot SM, Song H, Talbot WS, Tall GG, White JP, Wolfrum U, Xu L, Piao X. New functions and signaling mechanisms for the class of adhesion G protein-coupled receptors. Ann N Y Acad Sci 2014; 1333:43-64. [PMID: 25424900 DOI: 10.1111/nyas.12580] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The class of adhesion G protein-coupled receptors (aGPCRs), with 33 human homologs, is the second largest family of GPCRs. In addition to a seven-transmembrane α-helix-a structural feature of all GPCRs-the class of aGPCRs is characterized by the presence of a large N-terminal extracellular region. In addition, all aGPCRs but one (GPR123) contain a GPCR autoproteolysis-inducing (GAIN) domain that mediates autoproteolytic cleavage at the GPCR autoproteolysis site motif to generate N- and a C-terminal fragments (NTF and CTF, respectively) during protein maturation. Subsequently, the NTF and CTF are associated noncovalently as a heterodimer at the plasma membrane. While the biological function of the GAIN domain-mediated autocleavage is not fully understood, mounting evidence suggests that the NTF and CTF possess distinct biological activities in addition to their function as a receptor unit. We discuss recent advances in understanding the biological functions, signaling mechanisms, and disease associations of the aGPCRs.
Collapse
Affiliation(s)
- Ines Liebscher
- Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The zebrafish is a premier vertebrate model system that offers many experimental advantages for in vivo imaging and genetic studies. This review provides an overview of glial cell types in the central and peripheral nervous system of zebrafish. We highlight some recent work that exploited the strengths of the zebrafish system to increase the understanding of the role of Gpr126 in Schwann cell myelination and illuminate the mechanisms controlling oligodendrocyte development and myelination. We also summarize similarities and differences between zebrafish radial glia and mammalian astrocytes and consider the possibility that their distinct characteristics may represent extremes in a continuum of cell identity. Finally, we focus on the emergence of zebrafish as a model for elucidating the development and function of microglia. These recent studies have highlighted the power of the zebrafish system for analyzing important aspects of glial development and function.
Collapse
Affiliation(s)
- David A Lyons
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - William S Talbot
- Department of Developmental Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
40
|
Preston MA, Macklin WB. Zebrafish as a model to investigate CNS myelination. Glia 2014; 63:177-93. [PMID: 25263121 DOI: 10.1002/glia.22755] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/12/2014] [Indexed: 12/18/2022]
Abstract
Myelin plays a critical role in proper neuronal function by providing trophic and metabolic support to axons and facilitating energy-efficient saltatory conduction. Myelination is influenced by numerous molecules including growth factors, hormones, transmembrane receptors and extracellular molecules, which activate signaling cascades that drive cellular maturation. Key signaling molecules and downstream signaling cascades controlling myelination have been identified in cell culture systems. However, in vitro systems are not able to faithfully replicate the complex in vivo signaling environment that occurs during development or following injury. Currently, it remains time-consuming and expensive to investigate myelination in vivo in rodents, the most widely used model for studying mammalian myelination. As such, there is a need for alternative in vivo myelination models, particularly ones that can test molecular mechanisms without removing oligodendrocyte lineage cells from their native signaling environment or disrupting intercellular interactions with other cell types present during myelination. Here, we review the ever-increasing role of zebrafish in studies uncovering novel mechanisms controlling vertebrate myelination. These innovative studies range from observations of the behavior of single cells during in vivo myelination as well as mutagenesis- and pharmacology-based screens in whole animals. Additionally, we discuss recent efforts to develop novel models of demyelination and oligodendrocyte cell death in adult zebrafish for the study of cellular behavior in real time during repair and regeneration of damaged nervous systems.
Collapse
Affiliation(s)
- Marnie A Preston
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | | |
Collapse
|
41
|
The phosphate exporter xpr1b is required for differentiation of tissue-resident macrophages. Cell Rep 2014; 8:1659-1667. [PMID: 25220463 DOI: 10.1016/j.celrep.2014.08.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/23/2014] [Accepted: 08/08/2014] [Indexed: 12/23/2022] Open
Abstract
Phosphate concentration is tightly regulated at the cellular and organismal levels. The first metazoan phosphate exporter, XPR1, was recently identified, but its in vivo function remains unknown. In a genetic screen, we identified a mutation in a zebrafish ortholog of human XPR1, xpr1b. xpr1b mutants lack microglia, the specialized macrophages that reside in the brain, and also displayed an osteopetrotic phenotype characteristic of defects in osteoclast function. Transgenic expression studies indicated that xpr1b acts autonomously in developing macrophages. xpr1b mutants display no gross developmental defects that may arise from phosphate imbalance. We constructed a targeted mutation of xpr1a, a duplicate of xpr1b in the zebrafish genome, to determine whether Xpr1a and Xpr1b have redundant functions. Single mutants for xpr1a were viable, and double mutants for xpr1b;xpr1a were similar to xpr1b single mutants. Our genetic analysis reveals a specific role for the phosphate exporter Xpr1 in the differentiation of tissue macrophages.
Collapse
|
42
|
Salta E, Lau P, Sala Frigerio C, Coolen M, Bally-Cuif L, De Strooper B. A self-organizing miR-132/Ctbp2 circuit regulates bimodal notch signals and glial progenitor fate choice during spinal cord maturation. Dev Cell 2014; 30:423-36. [PMID: 25132384 DOI: 10.1016/j.devcel.2014.07.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 06/25/2014] [Accepted: 07/10/2014] [Indexed: 12/31/2022]
Abstract
Radial glial progenitors play pivotal roles in the development and patterning of the spinal cord, and their fate is controlled by Notch signaling. How Notch is shaped to regulate their crucial transition from expansion toward differentiation remains, however, unknown. miR-132 in the developing zebrafish dampens Notch signaling via a cascade involving the transcriptional corepressor Ctbp2 and the Notch suppressor Sirt1. At early embryonic stages, high Ctbp2 levels sustain Notch signaling and radial glial expansion and concomitantly induce miR-132 expression via a double-negative feedback loop involving Rest inhibition. The changing balance in miR-132 and Ctbp2 interaction gradually drives the switch in Notch output and radial glial progenitor fate as part of the larger developmental program involved in the transition from embryonic to larval spinal cord.
Collapse
Affiliation(s)
- Evgenia Salta
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics and Leuven Institute for Neurodegenerative Disorders, KU Leuven, 3000 Leuven, Belgium
| | - Pierre Lau
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics and Leuven Institute for Neurodegenerative Disorders, KU Leuven, 3000 Leuven, Belgium
| | - Carlo Sala Frigerio
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics and Leuven Institute for Neurodegenerative Disorders, KU Leuven, 3000 Leuven, Belgium
| | - Marion Coolen
- Zebrafish Neurogenetics Group, Laboratory of Neurobiology and Development, CNRS UPR 3294, Institute of Neurobiology Alfred Fessard, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cédex, France
| | - Laure Bally-Cuif
- Zebrafish Neurogenetics Group, Laboratory of Neurobiology and Development, CNRS UPR 3294, Institute of Neurobiology Alfred Fessard, Avenue de la Terrasse, 91198 Gif-sur-Yvette Cédex, France
| | - Bart De Strooper
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium; Center for Human Genetics and Leuven Institute for Neurodegenerative Disorders, KU Leuven, 3000 Leuven, Belgium; Institute of Neurology, University College London, Queen Square, WC1N 3BG London, UK.
| |
Collapse
|
43
|
Paavola KJ, Sidik H, Zuchero JB, Eckart M, Talbot WS. Type IV collagen is an activating ligand for the adhesion G protein-coupled receptor GPR126. Sci Signal 2014; 7:ra76. [PMID: 25118328 DOI: 10.1126/scisignal.2005347] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
GPR126 is an orphan heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptor (GPCR) that is essential for the development of diverse organs. We found that type IV collagen, a major constituent of the basement membrane, binds to Gpr126 and activates its signaling function. Type IV collagen stimulated the production of cyclic adenosine monophosphate in rodent Schwann cells, which require Gpr126 activity to differentiate, and in human embryonic kidney (HEK) 293 cells expressing exogenous Gpr126. Type IV collagen specifically bound to the extracellular amino-terminal region of Gpr126 containing the CUB (complement, Uegf, Bmp1) and pentraxin domains. Gpr126 derivatives lacking the entire amino-terminal region were constitutively active, suggesting that this region inhibits signaling and that ligand binding relieves this inhibition to stimulate receptor activity. A new zebrafish mutation that truncates Gpr126 after the CUB and pentraxin domains disrupted development of peripheral nerves and the inner ear. Thus, our findings identify type IV collagen as an activating ligand for GPR126, define its mechanism of activation, and highlight a previously unrecognized signaling function of type IV collagen in basement membranes.
Collapse
Affiliation(s)
- Kevin J Paavola
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Harwin Sidik
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - J Bradley Zuchero
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Eckart
- Protein and Nucleic Acid Facility, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - William S Talbot
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
44
|
Münzel EJ, Becker CG, Becker T, Williams A. Zebrafish regenerate full thickness optic nerve myelin after demyelination, but this fails with increasing age. Acta Neuropathol Commun 2014; 2:77. [PMID: 25022486 PMCID: PMC4164766 DOI: 10.1186/s40478-014-0077-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/19/2014] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION In the human demyelinating central nervous system (CNS) disease multiple sclerosis, remyelination promotes recovery and limits neurodegeneration, but this is inefficient and always ultimately fails. Furthermore, these regenerated myelin sheaths are thinner and shorter than the original, leaving the underlying axons potentially vulnerable. In rodent models, CNS remyelination is more efficient, so that in young animals (but not old) the number of myelinated axons is efficiently restored to normal, but in both young and old rodents, regenerated myelin sheaths are still short and thin. The reasons for these differences in remyelination efficiency, the thinner remyelinated myelin sheaths compared to developmental myelin and the subsequent effect on the underlying axon are unclear. We studied CNS remyelination in the highly regenerative adult zebrafish (Danio rerio), to better understand mechanisms of what we hypothesised would be highly efficient remyelination, and to identify differences to mammalian CNS remyelination, as larval zebrafish are increasingly used for high throughput screens to identify potential drug targets to improve myelination and remyelination. RESULTS We developed a novel method to induce a focal demyelinating lesion in adult zebrafish optic nerve with no discernible axonal damage, and describe the cellular changes over time. Remyelination is indeed efficient in both young and old adult zebrafish optic nerves, and at 4 weeks after demyelination, the number of myelinated axons is restored to normal, but internode lengths are short. However, unlike in rodents or in humans, in young zebrafish these regenerated myelin sheaths were of normal thickness, whereas in aged zebrafish, they were thin, and remained so even 3 months later. This inability to restore normal myelin thickness in remyelination with age was associated with a reduced macrophage/microglial response. CONCLUSION Zebrafish are able to efficiently restore normal thickness myelin around optic nerve axons after demyelination, unlike in mammals. However, this fails with age, when only thin myelin is achieved. This gives us a novel model to try and dissect the mechanism for restoring myelin thickness in CNS remyelination.
Collapse
Affiliation(s)
- Eva Jolanda Münzel
- />MRC Centre for Regenerative Medicine, SCRM, Edinburgh Bioquarter, 5 Little France Drive, Edinburgh, EH16 4UU UK
- />Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | | | - Thomas Becker
- />Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Anna Williams
- />MRC Centre for Regenerative Medicine, SCRM, Edinburgh Bioquarter, 5 Little France Drive, Edinburgh, EH16 4UU UK
| |
Collapse
|
45
|
Patra C, Monk KR, Engel FB. The multiple signaling modalities of adhesion G protein-coupled receptor GPR126 in development. ACTA ACUST UNITED AC 2014; 1:79. [PMID: 25493288 DOI: 10.14800/rci.79] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The G protein-coupled receptor (GPCR) superfamily is the largest known receptor family in the human genome. Although the family of adhesion GPCRs comprises the second largest sub-family, their function is poorly understood. Here, we review the current knowledge about the adhesion GPCR family member GPR126. GPR126 possesses a signal peptide, a 7TM domain homologous to secretin-like GPCRs, a GPS motif and an extended N-terminus containing a CUB (Complement, Uegf, Bmp1) domain, a PTX (Pentraxin) domain, a hormone binding domain and 27 putative N-glycosylation sites. Knockdown and knockout experiments in zebrafish and mice have demonstrated that Gpr126 plays an essential role in neural, cardiac and ear development. In addition, genome-wide association studies have implicated variations at the GPR126 locus in obstructive pulmonary dysfunction, in scoliosis and as a determinant of trunk length and body height. Gpr126 appears to exert its function depending on the organ system via G protein- and/or N-terminus-dependent signaling. Here, we review the current knowledge about Gpr126, which, due to the variety of its functions and its multiple signaling modalities, provides a model adhesion GPCR to understand general functional concepts utilized by adhesion GPCRs.
Collapse
Affiliation(s)
- Chinmoy Patra
- Department of Developmental Genetics, Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO 63110, USA
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, University of Erlangen-Nürnberg, Universitätsstraße 22, 91054 Erlangen, Germany
| |
Collapse
|
46
|
Abstract
INTRODUCTION Off-target effects represent one of the major concerns in the development of new pharmaceuticals, requiring large-scale animal toxicity testing. Faster, cheaper and more reliable assays based on zebrafish embryos (ZE) are being developed as major tools for assessing toxicity of chemicals during the drug-discovery process. AREAS COVERED This paper reviews techniques aimed to the analysis of in vivo sublethal toxic effects of drugs on major physiological functions, including the cardiovascular, nervous, neuromuscular, gastrointestinal and thyroid systems among others. Particular emphasis is placed on high-throughput screening techniques (HTS), including robotics, imaging technologies and image-analysis software. EXPERT OPINION The analysis of off-target effects of candidate drugs requires systemic analyses, as they often involve the complete organism rather than specific, tissue- or cell-specific targets. The unique physical and physiological characteristics of ZE make this system an essential tool for drug discovery and toxicity assessment. Different HTS methodologies applicable to ZE allow the screening of large numbers of different chemicals for many diverse and relevant toxic endpoints.
Collapse
Affiliation(s)
- Demetrio Raldúa
- IDAEA-CSIC, Environmental Chemistry , Jordi Girona 18, 08034 Barcelona , Spain +34 93400 6157 ; +34 93204 5904 ;
| | | |
Collapse
|
47
|
Shiau CE, Monk KR, Joo W, Talbot WS. An anti-inflammatory NOD-like receptor is required for microglia development. Cell Rep 2013; 5:1342-52. [PMID: 24316075 PMCID: PMC3878655 DOI: 10.1016/j.celrep.2013.11.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 09/05/2013] [Accepted: 11/04/2013] [Indexed: 12/31/2022] Open
Abstract
Microglia are phagocytic cells that form the basis of the brain's immune system. They derive from primitive macrophages that migrate into the brain during embryogenesis, but the genetic control of microglial development remains elusive. Starting with a genetic screen in zebrafish, we show that the noncanonical NOD-like receptor (NLR) nlrc3-like is essential for microglial formation. Although most NLRs trigger inflammatory signaling, nlrc3-like acts cell autonomously in microglia precursor cells to suppress unwarranted inflammation in the absence of overt immune challenge. In nlrc3-like mutants, primitive macrophages initiate a systemic inflammatory response with increased proinflammatory cytokines and actively aggregate instead of migrating into the brain to form microglia. NLRC3-like requires both its pyrin and NACHT domains, and it can bind the inflammasome component apoptosis-associated speck-like protein. Our studies suggest that NLRC3-like may regulate the inflammasome and other inflammatory pathways. Together, these results demonstrate that NLRC3-like prevents inappropriate macrophage activation, thereby allowing normal microglial development.
Collapse
Affiliation(s)
- Celia E Shiau
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Kelly R Monk
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - William Joo
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - William S Talbot
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
48
|
Glenn TD, Talbot WS. Signals regulating myelination in peripheral nerves and the Schwann cell response to injury. Curr Opin Neurobiol 2013; 23:1041-8. [PMID: 23896313 PMCID: PMC3830599 DOI: 10.1016/j.conb.2013.06.010] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 06/20/2013] [Accepted: 06/27/2013] [Indexed: 11/23/2022]
Abstract
In peripheral nerves, Schwann cells form myelin, which facilitates the rapid conduction of action potentials along axons in the vertebrate nervous system. Myelinating Schwann cells are derived from neural crest progenitors in a step-wise process that is regulated by extracellular signals and transcription factors. In addition to forming the myelin sheath, Schwann cells orchestrate much of the regenerative response that occurs after injury to peripheral nerves. In response to injury, myelinating Schwann cells dedifferentiate into repair cells that are essential for axonal regeneration, and then redifferentiate into myelinating Schwann cells to restore nerve function. Although this remarkable plasticity has long been recognized, many questions remain unanswered regarding the signaling pathways regulating both myelination and the Schwann cell response to injury.
Collapse
Affiliation(s)
- Thomas D. Glenn
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - William S. Talbot
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
49
|
Organ-specific function of adhesion G protein-coupled receptor GPR126 is domain-dependent. Proc Natl Acad Sci U S A 2013; 110:16898-903. [PMID: 24082093 DOI: 10.1073/pnas.1304837110] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite their abundance and multiple functions in a variety of organ systems, the function and signaling mechanisms of adhesion G protein-coupled receptors (GPCRs) are poorly understood. Adhesion GPCRs possess large N termini containing various functional domains. In addition, many of them are autoproteolytically cleaved at their GPS sites into an N-terminal fragment (NTF) and C-terminal fragment. Here we demonstrate that Gpr126 is expressed in the endocardium during early mouse heart development. Gpr126 knockout in mice and knockdown in zebrafish caused hypotrabeculation and affected mitochondrial function. Ectopic expression of Gpr126-NTF that lacks the GPS motif (NTF(ΔGPS)) in zebrafish rescued the trabeculation but not the previously described myelination phenotype in the peripheral nervous system. These data support a model in which the NTF of Gpr126, in contrast to the C-terminal fragment, plays an important role in heart development. Collectively, our analysis provides a unique example of the versatile function and signaling properties of adhesion GPCRs in vertebrates.
Collapse
|
50
|
Geng FS, Abbas L, Baxendale S, Holdsworth CJ, Swanson AG, Slanchev K, Hammerschmidt M, Topczewski J, Whitfield TT. Semicircular canal morphogenesis in the zebrafish inner ear requires the function of gpr126 (lauscher), an adhesion class G protein-coupled receptor gene. Development 2013; 140:4362-74. [PMID: 24067352 PMCID: PMC4007713 DOI: 10.1242/dev.098061] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Morphogenesis of the semicircular canal ducts in the vertebrate inner ear is a dramatic example of epithelial remodelling in the embryo, and failure of normal canal development results in vestibular dysfunction. In zebrafish and Xenopus, semicircular canal ducts develop when projections of epithelium, driven by extracellular matrix production, push into the otic vesicle and fuse to form pillars. We show that in the zebrafish, extracellular matrix gene expression is high during projection outgrowth and then rapidly downregulated after fusion. Enzymatic disruption of hyaluronan in the projections leads to their collapse and a failure to form pillars: as a result, the ears swell. We have cloned a zebrafish mutant, lauscher (lau), identified by its swollen ear phenotype. The primary defect in the ear is abnormal projection outgrowth and a failure of fusion to form the semicircular canal pillars. Otic expression of extracellular matrix components is highly disrupted: several genes fail to become downregulated and remain expressed at abnormally high levels into late larval stages. The lau mutations disrupt gpr126, an adhesion class G protein-coupled receptor gene. Expression of gpr126 is similar to that of sox10, an ear and neural crest marker, and is partially dependent on sox10 activity. Fusion of canal projections and downregulation of otic versican expression in a hypomorphic lau allele can be restored by cAMP agonists. We propose that Gpr126 acts through a cAMP-mediated pathway to control the outgrowth and adhesion of canal projections in the zebrafish ear via the regulation of extracellular matrix gene expression.
Collapse
Affiliation(s)
- Fan-Suo Geng
- MRC Centre for Developmental and Biomedical Genetics and Department of Biomedical Science, University of Sheffield, Sheffield, S10 2TN, UK
| | | | | | | | | | | | | | | | | |
Collapse
|