1
|
Balantzategi U, Gaminde-Blasco A, Kearns CA, Bayón-Cordero L, Sánchez-Gómez MV, Zugaza JL, Appel B, Alberdi E. Amyloid-β Dysregulates Oligodendroglial Lineage Cell Dynamics and Myelination via PKC in the Zebrafish Spinal Cord. Glia 2025. [PMID: 40087862 DOI: 10.1002/glia.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Soluble forms of amyloid-β (Aβ) peptide have been proposed as candidates to induce oligodendrocyte (OL) and myelin dysfunctions in the early stages of Alzheimer's disease (AD) pathology. Nevertheless, little is known about how Aβ affects OL differentiation and myelination in vivo, and the underlying molecular mechanisms. In this study, we explored the effects of a brain intraventricular injection of Aβ on OLs and myelin in the developing spinal cord of zebrafish larvae. Using quantitative fluorescent in situ RNA hybridization assays, we demonstrated that Aβ altered myrf and mbp mRNA levels and the regional distribution of mbp during larval development, suggesting an early differentiation of OLs. Through live imaging of Tg(myrf:mScarlet) and Tg(mbpa:tagRFP) zebrafish lines, both crossed with Tg(olig2:EGFP), we found that Aβ increased the number of myrf+ and mbp+ OLs in the dorsal spinal cord at 72 hpf and 5 dpf, respectively, without affecting total cell numbers. Furthermore, Aβ also increased the number of Sox10+cells, myelin sheaths per OL, and the number of myelinated axons in the dorsal spinal cord at 8 dpf compared to vehicle-injected control animals. Interestingly, the treatment of Aβ-injected zebrafish with the pan-PKC inhibitor Gö6983 restored the aforementioned alterations in OLs and myelin to control levels. Altogether, not only do we demonstrate that Aβ induces a precocious oligodendroglial differentiation leading to dysregulated myelination, but we also identified PKC as a key player in Aβ-induced pathology.
Collapse
Affiliation(s)
- Uxue Balantzategi
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Adhara Gaminde-Blasco
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Christina A Kearns
- Department of Pediatrics, Section of Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laura Bayón-Cordero
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - María Victoria Sánchez-Gómez
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - José Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain
| | - Bruce Appel
- Department of Pediatrics, Section of Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Elena Alberdi
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| |
Collapse
|
2
|
Henry R, Vander Heide R, Roy NM. Toxicity of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) on oligodendrocytes during embryonic zebrafish development. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 114:104627. [PMID: 39756717 DOI: 10.1016/j.etap.2025.104627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Polybrominated diphenyl ethers (PBDEs) are flame retardants heavily utilized across plastic, textile and electronic industries. Although these PBDEs are effective in protecting property and human life from fire, their high production volumes have led PBDEs to become pervasive environmental contaminants and pose an ecological and health risk as high levels have been noted in environmental media including water and sediment, wildlife and human tissue. Here we investigate the developmental neurotoxicity of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47), one of the more dominant PBDE congeners found in human tissue, on oligodendrocytes in the hindbrain and spinal cord. We utilized the zebrafish vertebrate model system and investigated low (5 µM) and high concentrations (20 µM) of BDE-47. We find that by 6 days post-fertilization, BDE-47 negatively affects oligodendrocyte development in the hindbrain and spinal cord in a concentration dependent manner.
Collapse
Affiliation(s)
- Ryann Henry
- Department of Biology, Sacred Heart University, Fairfield, CT, United States
| | - Reagan Vander Heide
- Department of Biology, Sacred Heart University, Fairfield, CT, United States
| | - Nicole M Roy
- Department of Biology, Sacred Heart University, Fairfield, CT, United States.
| |
Collapse
|
3
|
Walker WJ, Underwood KL, Garrett PI, Lorbacher KB, Linch SM, Rynes TP, Sloop C, Mruk K. Effects of age on the response to spinal cord injury: optimizing the larval zebrafish model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.05.18.541337. [PMID: 37292959 PMCID: PMC10245662 DOI: 10.1101/2023.05.18.541337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Zebrafish are an increasingly popular model to study regeneration after spinal cord injury (SCI). The transparency of larval zebrafish makes them ideal to study cellular processes in real time. Standardized approaches, including age at the time of injury, are not readily available making comparisons of the results with other models challenging. In this study, we systematically examined the response to spinal cord transection of larval zebrafish at three different larval ages (3-, 5-, or 7-days post fertilization (dpf)) to determine whether the developmental complexity of the larvae affects the overall response to SCI. We then used imaging and behavioral analysis to evaluate whether differences existed based on the age of injury. Injury led to increased expression of cytokines associated with the immune response; however, we found that the timing of specific inflammatory markers changed with the age of the injury. We also observed changes in glial and axonal bridging with age. Young larvae (3 dpf) were better able to regenerate axons independent of the glial bridge, unlike older larvae (7 dpf), consistent with results seen in adult zebrafish. Finally, locomotor experiments demonstrated that some swimming behavior occurs independent of glial bridge formation, further highlighting the need for standardization of this model and functional recovery assays. Overall, we found differences based on the age of transection in larval zebrafish, underlining the importance of considering age when designing experiments aimed at understanding regeneration.
Collapse
|
4
|
Nabizadeh F. Brain white matter damage biomarkers. Adv Clin Chem 2024; 125:55-91. [PMID: 39988408 DOI: 10.1016/bs.acc.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
White matter (WM), constituting nearly half of the human brain's mass, is pivotal for the rapid transmission of neural signals across different brain regions, significantly influencing cognitive processes like learning, memory, and problem-solving. The integrity of WM is essential for brain function, and its damage, which can occur due to conditions such as multiple sclerosis (MS), stroke, and traumatic brain injury, results in severe neurological deficits and cognitive decline. The primary objective of this book chapter is to discuss the clinical significance of fluid biomarkers in assessing WM damage within the central nervous system (CNS). It explores the biological underpinnings and pathological changes in WM due to various neurological conditions and details how alterations can be detected and quantified through fluid biomarkers. By examining biomarkers like Myelin Basic Protein (MBP), Neurofilament light chain (NFL), and others, the chapter highlights their role in enhancing diagnostic precision, monitoring disease progression, and guiding therapeutic interventions, thus providing crucial insights into maintaining WM integrity and preventing cognitive and physical disabilities.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- School of Medicine, Iran University of Medical Sciences, and Alzheimer's Disease Institute, Tehran, Iran.
| |
Collapse
|
5
|
Czopka T, Monk K, Peri F. Glial Cell Development and Function in the Zebrafish Central Nervous System. Cold Spring Harb Perspect Biol 2024; 16:a041350. [PMID: 38692835 PMCID: PMC11529855 DOI: 10.1101/cshperspect.a041350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Over the past decades the zebrafish has emerged as an excellent model organism with which to study the biology of all glial cell types in nervous system development, plasticity, and regeneration. In this review, which builds on the earlier work by Lyons and Talbot in 2015, we will summarize how the relative ease to manipulate the zebrafish genome and its suitability for intravital imaging have helped understand principles of glial cell biology with a focus on oligodendrocytes, microglia, and astrocytes. We will highlight recent findings on the diverse properties and functions of these glial cell types in the central nervous system and discuss open questions and future directions of the field.
Collapse
Affiliation(s)
- Tim Czopka
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Kelly Monk
- Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Francesca Peri
- Department of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
6
|
Maktabi B, Collins A, Safee R, Bouyer J, Wisner AS, Williams FE, Schiefer IT. Zebrafish as a Model for Multiple Sclerosis. Biomedicines 2024; 12:2354. [PMID: 39457666 PMCID: PMC11504653 DOI: 10.3390/biomedicines12102354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Zebrafish have become a key model organism in neuroscience research because of their unique advantages. Their genetic, anatomical, and physiological similarities to humans, coupled with their rapid development and transparent embryos, make them an excellent tool for investigating various aspects of neurobiology. They have specifically emerged as a valuable and versatile model organism in biomedical research, including the study of neurological disorders such as multiple sclerosis. Multiple sclerosis is a chronic autoimmune disease known to cause damage to the myelin sheath that protects the nerves in the brain and spinal cord. Objective: This review emphasizes the importance of continued research in both in vitro and in vivo models to advance our understanding of MS and develop effective treatments, ultimately improving the quality of life for those affected by this debilitating disease. Conclusions: Recent studies show the significance of zebrafish as a model organism for investigating demyelination and remyelination processes, providing new insights into MS pathology and potential therapies.
Collapse
Affiliation(s)
- Briana Maktabi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Abigail Collins
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Raihaanah Safee
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
| | - Jada Bouyer
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Alexander S. Wisner
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Frederick E. Williams
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Isaac T. Schiefer
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
7
|
Filippini A, Cannone E, Mazziotti V, Carini G, Mutti V, Ravelli C, Gennarelli M, Schiavone M, Russo I. Leucine-Rich Repeat Kinase-2 Controls the Differentiation and Maturation of Oligodendrocytes in Mice and Zebrafish. Biomolecules 2024; 14:870. [PMID: 39062584 PMCID: PMC11274935 DOI: 10.3390/biom14070870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Leucine-rich repeat kinase-2 (LRRK2), a gene mutated in familial and sporadic Parkinson's disease (PD), controls multiple cellular processes important for GLIA physiology. Interestingly, emerging studies report that LRRK2 is highly expressed in oligodendrocyte precursor cells (OPCs) compared to the pathophysiology of other brain cells and oligodendrocytes (OLs) in PD. Altogether, these observations suggest crucial function(s) of LRRK2 in OPCs/Ols, which would be interesting to explore. In this study, we investigated the role of LRRK2 in OLs. We showed that LRRK2 knock-out (KO) OPC cultures displayed defects in the transition of OPCs into OLs, suggesting a role of LRRK2 in OL differentiation. Consistently, we found an alteration of myelin basic protein (MBP) striosomes in LRRK2 KO mouse brains and reduced levels of oligodendrocyte transcription factor 2 (Olig2) and Mbp in olig2:EGFP and mbp:RFP transgenic zebrafish embryos injected with lrrk2 morpholino (MO). Moreover, lrrk2 knock-down zebrafish exhibited a lower amount of nerve growth factor (Ngf) compared to control embryos, which represents a potent regulator of oligodendrogenesis and myelination. Overall, our findings indicate that LRRK2 controls OL differentiation, affecting the number of mature OLs.
Collapse
Affiliation(s)
- Alice Filippini
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.F.); (E.C.); (G.C.); (M.G.)
| | - Elena Cannone
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.F.); (E.C.); (G.C.); (M.G.)
| | - Valentina Mazziotti
- IRCCS Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.); (V.M.)
| | - Giulia Carini
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.F.); (E.C.); (G.C.); (M.G.)
| | - Veronica Mutti
- IRCCS Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.); (V.M.)
| | - Cosetta Ravelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Massimo Gennarelli
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.F.); (E.C.); (G.C.); (M.G.)
- IRCCS Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.); (V.M.)
| | - Marco Schiavone
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.F.); (E.C.); (G.C.); (M.G.)
| | - Isabella Russo
- Unit of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (A.F.); (E.C.); (G.C.); (M.G.)
- IRCCS Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy; (V.M.); (V.M.)
| |
Collapse
|
8
|
Rutherford HA, Candeias D, Duncan CJA, Renshaw SA, Hamilton N. Macrophage transplantation rescues RNASET2-deficient leukodystrophy by replacing deficient microglia in a zebrafish model. Proc Natl Acad Sci U S A 2024; 121:e2321496121. [PMID: 38753517 PMCID: PMC11126979 DOI: 10.1073/pnas.2321496121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
RNASET2-deficient leukodystrophy is a rare infantile white matter disorder mimicking a viral infection and resulting in severe psychomotor impairments. Despite its severity, there is little understanding of cellular mechanisms of pathogenesis and no treatments. Recent research using the rnaset2 mutant zebrafish model has suggested that microglia may be the drivers of the neuropathology, due to their failure to digest apoptotic debris during neurodevelopment. Therefore, we developed a strategy for microglial replacement through transplantation of adult whole kidney marrow-derived macrophages into embryonic hosts. Using live imaging, we revealed that transplant-derived macrophages can engraft within host brains and express microglia-specific markers, suggesting the adoption of a microglial phenotype. Tissue-clearing strategies revealed the persistence of transplanted cells in host brains beyond embryonic stages. We demonstrated that transplanted cells clear apoptotic cells within the brain, as well as rescue overactivation of the antiviral response otherwise seen in mutant larvae. RNA sequencing at the point of peak transplant-derived cell engraftment confirms that transplantation can reduce the brain-wide immune response and particularly, the antiviral response, in rnaset2-deficient brains. Crucially, this reduction in neuroinflammation resulted in behavioral rescue-restoring rnaset2 mutant motor activity to wild-type (WT) levels in embryonic and juvenile stages. Together, these findings demonstrate the role of microglia as the cellular drivers of neuropathology in rnaset2 mutants and that macrophage transplantation is a viable strategy for microglial replacement in the zebrafish. Therefore, microglia-targeted interventions may have therapeutic benefits in RNASET2-deficient leukodystrophy.
Collapse
Affiliation(s)
- Holly A. Rutherford
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Diogo Candeias
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| | - Christopher J. A. Duncan
- Immunology and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, NewcastleNE2 4HH, United Kingdom
- Department of Infection and Tropical Medicine, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals National Health Services Foundation Trust, NewcastleNE2 4HH, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Noémie Hamilton
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| |
Collapse
|
9
|
Bouchard EL, Meireles AM, Talbot WS. Oligodendrocyte development and myelin sheath formation are regulated by the antagonistic interaction between the Rag-Ragulator complex and TFEB. Glia 2024; 72:289-299. [PMID: 37767930 PMCID: PMC10841052 DOI: 10.1002/glia.24473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 08/11/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023]
Abstract
Myelination by oligodendrocytes is critical for fast axonal conduction and for the support and survival of neurons in the central nervous system. Recent studies have emphasized that myelination is plastic and that new myelin is formed throughout life. Nonetheless, the mechanisms that regulate the number, length, and location of myelin sheaths formed by individual oligodendrocytes are incompletely understood. Previous work showed that the lysosomal transcription factor TFEB represses myelination by oligodendrocytes and that the RagA GTPase inhibits TFEB, but the step or steps of myelination in which TFEB plays a role have remained unclear. Here, we show that TFEB regulates oligodendrocyte differentiation and also controls the length of myelin sheaths formed by individual oligodendrocytes. In the dorsal spinal cord of tfeb mutants, individual oligodendrocytes produce myelin sheaths that are longer than those produced by wildtype cells. Transmission electron microscopy shows that there are more myelinated axons in the dorsal spinal cord of tfeb mutants than in wildtype animals, but no significant change in axon diameter. In contrast to tfeb mutants, oligodendrocytes in rraga mutants produce shorter myelin sheaths. The sheath length in rraga; tfeb double mutants is not significantly different from wildtype, consistent with the antagonistic interaction between RagA and TFEB. Finally, we find that the GTPase activating protein Flcn and the RagCa and RagCb GTPases are also necessary for myelination by oligodendrocytes. These findings demonstrate that TFEB coordinates myelin sheath length and number during myelin formation in the central nervous system.
Collapse
Affiliation(s)
- Ellen L. Bouchard
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ana M. Meireles
- 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
|
10
|
Shih HY, Raas Q, Bonkowsky JL. Progress in leukodystrophies with zebrafish. Dev Growth Differ 2024; 66:21-34. [PMID: 38239149 DOI: 10.1111/dgd.12907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/31/2024]
Abstract
Inherited leukodystrophies are genetic disorders characterized by abnormal white matter in the central nervous system. Although individually rare, there are more than 400 distinct types of leukodystrophies with a cumulative incidence of 1 in 4500 live births. The pathophysiology of most leukodystrophies is poorly understood, there are treatments for only a few, and there is significant morbidity and mortality, suggesting a critical need for improvements in this field. A variety of animal, cell, and induced pluripotent stem cell-derived models have been developed for leukodystrophies, but with significant limitations in all models. Many leukodystrophies lack animal models, and extant models often show no or mixed recapitulation of key phenotypes. Zebrafish (Danio rerio) have become increasingly used as disease models for studying leukodystrophies due to their early onset of disease phenotypes and conservation of molecular and neurobiological mechanisms. Here, we focus on reviewing new zebrafish disease models for leukodystrophy or models with recent progress. This includes discussion of leukodystrophy with vanishing white matter disease, X-linked adrenoleukodystrophy, Zellweger spectrum disorders and peroxisomal disorders, PSAP deficiency, metachromatic leukodystrophy, Krabbe disease, hypomyelinating leukodystrophy-8/4H leukodystrophy, Aicardi-Goutières syndrome, RNASET2-deficient cystic leukoencephalopathy, hereditary diffuse leukoencephalopathy with spheroids-1 (CSF1R-related leukoencephalopathy), and ultra-rare leukodystrophies. Zebrafish models offer important potentials for the leukodystrophy field, including testing of new variants in known genes; establishing causation of newly discovered genes; and early lead compound identification for therapies. There are also unrealized opportunities to use humanized zebrafish models which have been sparsely explored.
Collapse
Affiliation(s)
- Hung-Yu Shih
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biological Sciences, Utah Tech University, Saint George, Utah, USA
- Center for Precision & Functional Genomics, Utah Tech University, Saint George, Utah, USA
| | - Quentin Raas
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM UMR 1163, Paris, France
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Center for Personalized Medicine, Primary Children's Hospital, Salt Lake City, Utah, USA
| |
Collapse
|
11
|
Shi L, Wang Z, Li Y, Song Z, Yin W, Hu B. Deletion of the chd7 Hinders Oligodendrocyte Progenitor Cell Development and Myelination in Zebrafish. Int J Mol Sci 2023; 24:13535. [PMID: 37686337 PMCID: PMC10488005 DOI: 10.3390/ijms241713535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
CHD7, an encoding ATP-dependent chromodomain helicase DNA-binding protein 7, has been identified as the causative gene involved in CHARGE syndrome (Coloboma of the eye, Heart defects, Atresia choanae, Retardation of growth and/or development, Genital abnormalities and Ear abnormalities). Although studies in rodent models have expanded our understanding of CHD7, its role in oligodendrocyte (OL) differentiation and myelination in zebrafish is still unclear. In this study, we generated a chd7-knockout strain with CRISPR/Cas9 in zebrafish. We observed that knockout (KO) of chd7 intensely impeded the oligodendrocyte progenitor cells' (OPCs) migration and myelin formation due to massive expression of chd7 in oilg2+ cells, which might provoke upregulation of the MAPK signal pathway. Thus, our study demonstrates that chd7 is critical to oligodendrocyte migration and myelination during early development in zebrafish and describes a mechanism potentially associated with CHARGE syndrome.
Collapse
Affiliation(s)
- Lingyu Shi
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (L.S.)
| | - Zongyi Wang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (L.S.)
| | - Yujiao Li
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (L.S.)
| | - Zheng Song
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (L.S.)
| | - Wu Yin
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Bing Hu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (L.S.)
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| |
Collapse
|
12
|
Kalotay E, Klugmann M, Housley GD, Fröhlich D. Recessive aminoacyl-tRNA synthetase disorders: lessons learned from in vivo disease models. Front Neurosci 2023; 17:1182874. [PMID: 37274208 PMCID: PMC10234152 DOI: 10.3389/fnins.2023.1182874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
Protein synthesis is a fundamental process that underpins almost every aspect of cellular functioning. Intriguingly, despite their common function, recessive mutations in aminoacyl-tRNA synthetases (ARSs), the family of enzymes that pair tRNA molecules with amino acids prior to translation on the ribosome, cause a diverse range of multi-system disorders that affect specific groups of tissues. Neurological development is impaired in most ARS-associated disorders. In addition to central nervous system defects, diseases caused by recessive mutations in cytosolic ARSs commonly affect the liver and lungs. Patients with biallelic mutations in mitochondrial ARSs often present with encephalopathies, with variable involvement of peripheral systems. Many of these disorders cause severe disability, and as understanding of their pathogenesis is currently limited, there are no effective treatments available. To address this, accurate in vivo models for most of the recessive ARS diseases are urgently needed. Here, we discuss approaches that have been taken to model recessive ARS diseases in vivo, highlighting some of the challenges that have arisen in this process, as well as key results obtained from these models. Further development and refinement of animal models is essential to facilitate a better understanding of the pathophysiology underlying recessive ARS diseases, and ultimately to enable development and testing of effective therapies.
Collapse
Affiliation(s)
- Elizabeth Kalotay
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gary D. Housley
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Dominik Fröhlich
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
13
|
Choi EK, Choi BM, Cho Y, Kim S. Myelin toxicity of chlorhexidine in zebrafish larvae. Pediatr Res 2023; 93:845-851. [PMID: 35854088 DOI: 10.1038/s41390-022-02186-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/27/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Chlorhexidine gluconate (CHG) is a topical antiseptic solution recommended for skin preparation before central venous catheter placement and maintenance in adults and children. Although CHG is not recommended for use in children aged <2 months owing to limited safety data, it is commonly used in neonatal intensive care units worldwide. We used zebrafish model to verify the effects of early-life exposure to CHG on the developing nervous system, highlighting its impact on oligodendrocyte development and myelination. METHODS Zebrafish embryos were exposed to different concentrations of CHG from 4 h post fertilization to examine developmental toxicity. The hatching rate, mortality, and malformation of the embryos/larvae were monitored. Oligodendrocyte lineage in transgenic zebrafish embryos was used to investigate defects in oligodendrocytes and myelin. Myelin structure, locomotor behavior, and expression levels of genes involved in myelination were investigated. RESULTS Exposure to CHG significantly induced oligodendrocyte defects in the central nervous system, delayed myelination, and locomotor alterations. Ultra-microstructural changes with splitting and fluid-accumulated vacuoles between the myelin sheaths were found. Embryonic exposure to CHG decreased myelination, in association with downregulated mbpa, plp1b, and scrt2 gene expression. CONCLUSION Our results suggest that CHG has a potential for myelin toxicity in the developing brain. IMPACT To date, the neurodevelopmental toxicity of chlorhexidine gluconate (CHG) exposure on the developing brains of infants remains unknown. We demonstrated that CHG exposure to zebrafish larvae resulted in significant defects in oligodendrocytes and myelin sheaths. These CHG-exposed zebrafish larvae exhibited structural changes and locomotor alterations. Given the increased CHG use in neonates, this study is the first to identify the risk of early-life CHG exposure on the developing nervous system.
Collapse
Affiliation(s)
- Eui Kyung Choi
- Department of Pediatrics, College of Medicine, Korea University, Seoul, Republic of Korea
- Division of Neonatology, Department of Pediatrics, Korea University Guro Hospital, Ulsan, Gyeonggi-do, Republic of Korea
| | - Byung Min Choi
- Department of Pediatrics, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yuji Cho
- Core Research & Development Center, Korea University Ansan Hospital, Ansan, Gyeonggi-do, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Suhyun Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Republic of Korea.
- Zebrafish Translational Medical Research Center, Korea University, Ansan, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
14
|
Santos-Ledo A, Pérez-Montes C, DeOliveira-Mello L, Arévalo R, Velasco A. Oligodendrocyte origin and development in the zebrafish visual system. J Comp Neurol 2023; 531:515-527. [PMID: 36477827 PMCID: PMC10107312 DOI: 10.1002/cne.25440] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 09/19/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Oligodendrocytes are the myelinating cells in the central nervous system. In birds and mammals, the oligodendrocyte progenitor cells (OPCs) originate in the preoptic area (POA) of the hypothalamus. However, it remains unclear in other vertebrates such as fish. Thus, we have studied the early progression of OPCs during zebrafish visual morphogenesis from 2 days post fertilization (dpf) until 11 dpf using the olig2:EGFP transgenic line; and we have analyzed the differential expression of transcription factors involved in oligodendrocyte differentiation: Sox2 (using immunohistochemistry) and Sox10 (using the transgenic line sox10:tagRFP). The first OPCs (olig2:EGFP/Sox2) were found at 2 dpf in the POA. From 3 dpf onwards, these olig2:EGFP/Sox2 cells migrate to the optic chiasm, where they invade the optic nerve (ON), extending toward the retina. At 5 dpf, olig2:EGFP/Sox2 cells in the ON also colocalize with sox10:tagRFP. When olig2:EGFP cells differentiate and present more projections, they become positive only for sox10:tagRFP. olig2:EGFP/sox10: tagRFP cells ensheath the ON by 5 dpf when they also become positive for a myelin marker, based on the mbpa:tagRFPt transgenic line. We also found olig2:EGFP cells in other regions of the visual system. In the central retina at 2 dpf, they are positive for Sox2 but later become restricted to the proliferative germinal zone without this marker. In the ventricular areas of the optic tectum, olig2:EGFP cells present Sox2 but arborized ones sox10:tagRFP instead. Our data matches with other models, where OPCs are specified in the POA and migrate to the ON through the optic chiasm.
Collapse
Affiliation(s)
- Adrián Santos-Ledo
- Department of Cell Biology and Pathology, Instituto de NeurocienciasdeCastilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Cristina Pérez-Montes
- Department of Cell Biology and Pathology, Instituto de NeurocienciasdeCastilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Laura DeOliveira-Mello
- Department of Cell Biology and Pathology, Instituto de NeurocienciasdeCastilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Rosario Arévalo
- Department of Cell Biology and Pathology, Instituto de NeurocienciasdeCastilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Almudena Velasco
- Department of Cell Biology and Pathology, Instituto de NeurocienciasdeCastilla y León (INCyL), Universidad de Salamanca, Salamanca, Spain.,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| |
Collapse
|
15
|
Shoenhard H, Jain RA, Granato M. The calcium-sensing receptor (CaSR) regulates zebrafish sensorimotor decision making via a genetically defined cluster of hindbrain neurons. Cell Rep 2022; 41:111790. [PMID: 36476852 PMCID: PMC9813870 DOI: 10.1016/j.celrep.2022.111790] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Decision making is a fundamental nervous system function that ranges widely in complexity and speed of execution. We previously established larval zebrafish as a model for sensorimotor decision making and identified the G-protein-coupled calcium-sensing receptor (CaSR) to be critical for this process. Here, we report that CaSR functions in neurons to dynamically regulate the bias between two behavioral outcomes: escapes and reorientations. By employing a computational guided transgenic strategy, we identify a genetically defined neuronal cluster in the hindbrain as a key candidate site for CaSR function. Finally, we demonstrate that transgenic CaSR expression targeting this cluster consisting of a few hundred neurons shifts behavioral bias in wild-type animals and restores decision making deficits in CaSR mutants. Combined, our data provide a rare example of a G-protein-coupled receptor that biases vertebrate sensorimotor decision making via a defined neuronal cluster.
Collapse
Affiliation(s)
- Hannah Shoenhard
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roshan A Jain
- Department of Biology, Haverford College, Haverford, PA 19041, USA
| | - Michael Granato
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Spencer SA, Suárez-Pozos E, Verdugo JS, Wang H, Afshari FS, Li G, Manam S, Yasuda D, Ortega A, Lister JA, Ishii S, Zhang Y, Fuss B. Lysophosphatidic acid signaling via LPA 6 : A negative modulator of developmental oligodendrocyte maturation. J Neurochem 2022; 163:478-499. [PMID: 36153691 PMCID: PMC9772207 DOI: 10.1111/jnc.15696] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/14/2023]
Abstract
The developmental process of central nervous system (CNS) myelin sheath formation is characterized by well-coordinated cellular activities ultimately ensuring rapid and synchronized neural communication. During this process, myelinating CNS cells, namely oligodendrocytes (OLGs), undergo distinct steps of differentiation, whereby the progression of earlier maturation stages of OLGs represents a critical step toward the timely establishment of myelinated axonal circuits. Given the complexity of functional integration, it is not surprising that OLG maturation is controlled by a yet fully to be defined set of both negative and positive modulators. In this context, we provide here first evidence for a role of lysophosphatidic acid (LPA) signaling via the G protein-coupled receptor LPA6 as a negative modulatory regulator of myelination-associated gene expression in OLGs. More specifically, the cell surface accessibility of LPA6 was found to be restricted to the earlier maturation stages of differentiating OLGs, and OLG maturation was found to occur precociously in Lpar6 knockout mice. To further substantiate these findings, a novel small molecule ligand with selectivity for preferentially LPA6 and LPA6 agonist characteristics was functionally characterized in vitro in primary cultures of rat OLGs and in vivo in the developing zebrafish. Utilizing this approach, a negative modulatory role of LPA6 signaling in OLG maturation could be corroborated. During development, such a functional role of LPA6 signaling likely serves to ensure timely coordination of circuit formation and myelination. Under pathological conditions as seen in the major human demyelinating disease multiple sclerosis (MS), however, persistent LPA6 expression and signaling in OLGs can be seen as an inhibitor of myelin repair. Thus, it is of interest that LPA6 protein levels appear elevated in MS brain samples, thereby suggesting that LPA6 signaling may represent a potential new druggable pathway suitable to promote myelin repair in MS.
Collapse
Affiliation(s)
- Samantha A Spencer
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Edna Suárez-Pozos
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Jazmín Soto Verdugo
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - Huiqun Wang
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Fatemah S Afshari
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Guo Li
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Susmita Manam
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Daisuke Yasuda
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Arturo Ortega
- Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de México, México
| | - James A Lister
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Satoshi Ishii
- Department of Immunology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| |
Collapse
|
17
|
Humanized zebrafish as a tractable tool for in vivo evaluation of pro-myelinating drugs. Cell Chem Biol 2022; 29:1541-1555.e7. [PMID: 36126653 DOI: 10.1016/j.chembiol.2022.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/25/2022] [Accepted: 08/24/2022] [Indexed: 12/14/2022]
Abstract
Therapies that promote neuroprotection and axonal survival by enhancing myelin regeneration are an unmet need to prevent disability progression in multiple sclerosis. Numerous potentially beneficial compounds have originated from phenotypic screenings but failed in clinical trials. It is apparent that current cell- and animal-based disease models are poor predictors of positive treatment options, arguing for novel experimental approaches. Here we explore the experimental power of humanized zebrafish to foster the identification of pro-remyelination compounds via specific inhibition of GPR17. Using biochemical and imaging techniques, we visualize the expression of zebrafish (zf)-gpr17 during the distinct stages of oligodendrocyte development, thereby demonstrating species-conserved expression between zebrafish and mammals. We also demonstrate species-conserved function of zf-Gpr17 using genetic loss-of-function and rescue techniques. Finally, using GPR17-humanized zebrafish, we provide proof of principle for in vivo analysis of compounds acting via targeted inhibition of human GPR17. We anticipate that GPR17-humanized zebrafish will markedly improve the search for effective pro-myelinating pharmacotherapies.
Collapse
|
18
|
Auer F, Schoppik D. The Larval Zebrafish Vestibular System Is a Promising Model to Understand the Role of Myelin in Neural Circuits. Front Neurosci 2022; 16:904765. [PMID: 35600621 PMCID: PMC9122096 DOI: 10.3389/fnins.2022.904765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/11/2022] [Indexed: 12/27/2022] Open
Abstract
Myelin is classically known for its role in facilitating nerve conduction. However, recent work casts myelin as a key player in both proper neuronal circuit development and function. With this expanding role comes a demand for new approaches to characterize and perturb myelin in the context of tractable neural circuits as they mature. Here we argue that the simplicity, strong conservation, and clinical relevance of the vestibular system offer a way forward. Further, the tractability of the larval zebrafish affords a uniquely powerful means to test open hypotheses of myelin's role in normal development and disordered vestibular circuits. We end by identifying key open questions in myelin neurobiology that the zebrafish vestibular system is particularly well-suited to address.
Collapse
Affiliation(s)
| | - David Schoppik
- Departments of Otolaryngology, Neuroscience & Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
19
|
Carvalho E, Morais M, Ferreira H, Silva M, Guimarães S, Pêgo A. A paradigm shift: Bioengineering meets mechanobiology towards overcoming remyelination failure. Biomaterials 2022; 283:121427. [DOI: 10.1016/j.biomaterials.2022.121427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 01/31/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022]
|
20
|
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: 9] [Impact Index Per Article: 2.3] [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
|
21
|
Small molecule screening as an approach to encounter inefficient myelin repair. Curr Opin Pharmacol 2021; 61:127-135. [PMID: 34753035 DOI: 10.1016/j.coph.2021.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/28/2021] [Accepted: 09/30/2021] [Indexed: 11/20/2022]
Abstract
While current multiple sclerosis therapies are focused on immunomodulation, thereby slowing down disease progression, scientific interest has nowadays been shifted toward regenerative therapies aiming at reversing already existing deficits. The application of chemical compounds was proven to be valuable for the understanding of oligodendrogenesis and for exposing mechanisms that can boost remyelination. However, sufficient myelin repair has not been achieved yet, thus underscoring the need for more studies toward this unmet clinical goal. In this regard, many research groups have significantly contributed to the field via developing compound screening approaches or using single substances. We, here, present an overview of recent studies addressing the identification of myelin repair drugs and provide insights into technical aspects and identified substances.
Collapse
|
22
|
Madden ME, Suminaite D, Ortiz E, Early JJ, Koudelka S, Livesey MR, Bianco IH, Granato M, Lyons DA. CNS Hypomyelination Disrupts Axonal Conduction and Behavior in Larval Zebrafish. J Neurosci 2021; 41:9099-9111. [PMID: 34544838 PMCID: PMC8570833 DOI: 10.1523/jneurosci.0842-21.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 11/21/2022] Open
Abstract
Myelination is essential for central nervous system (CNS) formation, health and function. As a model organism, larval zebrafish have been extensively employed to investigate the molecular and cellular basis of CNS myelination, because of their genetic tractability and suitability for non-invasive live cell imaging. However, it has not been assessed to what extent CNS myelination affects neural circuit function in zebrafish larvae, prohibiting the integration of molecular and cellular analyses of myelination with concomitant network maturation. To test whether larval zebrafish might serve as a suitable platform with which to study the effects of CNS myelination and its dysregulation on circuit function, we generated zebrafish myelin regulatory factor (myrf) mutants with CNS-specific hypomyelination and investigated how this affected their axonal conduction properties and behavior. We found that myrf mutant larvae exhibited increased latency to perform startle responses following defined acoustic stimuli. Furthermore, we found that hypomyelinated animals often selected an impaired response to acoustic stimuli, exhibiting a bias toward reorientation behavior instead of the stimulus-appropriate startle response. To begin to study how myelination affected the underlying circuitry, we established electrophysiological protocols to assess various conduction properties along single axons. We found that the hypomyelinated myrf mutants exhibited reduced action potential conduction velocity and an impaired ability to sustain high-frequency action potential firing. This study indicates that larval zebrafish can be used to bridge molecular and cellular investigation of CNS myelination with multiscale assessment of neural circuit function.SIGNIFICANCE STATEMENT Myelination of CNS axons is essential for their health and function, and it is now clear that myelination is a dynamic life-long process subject to modulation by neuronal activity. However, it remains unclear precisely how changes to myelination affects animal behavior and underlying action potential conduction along axons in intact neural circuits. In recent years, zebrafish have been employed to study cellular and molecular mechanisms of myelination, because of their relatively simple, optically transparent, experimentally tractable vertebrate nervous system. Here we find that changes to myelination alter the behavior of young zebrafish and action potential conduction along individual axons, providing a platform to integrate molecular, cellular, and circuit level analyses of myelination using this model.
Collapse
Affiliation(s)
- M E Madden
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - D Suminaite
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - E Ortiz
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - J J Early
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - S Koudelka
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - M R Livesey
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, The University of Sheffield, Sheffield S10 2HQ, United Kingdom
| | - I H Bianco
- Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - M Granato
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - D A Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| |
Collapse
|
23
|
Farías-Serratos BM, Lazcano I, Villalobos P, Darras VM, Orozco A. Thyroid hormone deficiency during zebrafish development impairs central nervous system myelination. PLoS One 2021; 16:e0256207. [PMID: 34403440 PMCID: PMC8370640 DOI: 10.1371/journal.pone.0256207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 08/02/2021] [Indexed: 11/19/2022] Open
Abstract
Thyroid hormones are messengers that bind to specific nuclear receptors and regulate a wide range of physiological processes in the early stages of vertebrate embryonic development, including neurodevelopment and myelogenesis. We here tested the effects of reduced T3 availability upon the myelination process by treating zebrafish embryos with low concentrations of iopanoic acid (IOP) to block T4 to T3 conversion. Black Gold II staining showed that T3 deficiency reduced the myelin density in the forebrain, midbrain, hindbrain and the spinal cord at 3 and 7 dpf. These observations were confirmed in 3 dpf mbp:egfp transgenic zebrafish, showing that the administration of IOP reduced the fluorescent signal in the brain. T3 rescue treatment restored brain myelination and reversed the changes in myelin-related gene expression induced by IOP exposure. NG2 immunostaining revealed that T3 deficiency reduced the amount of oligodendrocyte precursor cells in 3 dpf IOP-treated larvae. Altogether, the present results show that inhibition of T4 to T3 conversion results in hypomyelination, suggesting that THs are part of the key signaling molecules that control the timing of oligodendrocyte differentiation and myelin synthesis from very early stages of brain development.
Collapse
Affiliation(s)
| | - Iván Lazcano
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
| | - Patricia Villalobos
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
| | - Veerle M. Darras
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
- Biology Department, Laboratory of Comparative Endocrinology, KU Leuven, Leuven, Belgium
| | - Aurea Orozco
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Qro., México
- * E-mail:
| |
Collapse
|
24
|
Berdowski WM, Sanderson LE, van Ham TJ. The multicellular interplay of microglia in health and disease: lessons from leukodystrophy. Dis Model Mech 2021; 14:dmm048925. [PMID: 34282843 PMCID: PMC8319551 DOI: 10.1242/dmm.048925] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Microglia are highly dynamic cells crucial for developing and maintaining lifelong brain function and health through their many interactions with essentially all cellular components of the central nervous system. The frequent connection of microglia to leukodystrophies, genetic disorders of the white matter, has highlighted their involvement in the maintenance of white matter integrity. However, the mechanisms that underlie their putative roles in these processes remain largely uncharacterized. Microglia have also been gaining attention as possible therapeutic targets for many neurological conditions, increasing the demand to understand their broad spectrum of functions and the impact of their dysregulation. In this Review, we compare the pathological features of two groups of genetic leukodystrophies: those in which microglial dysfunction holds a central role, termed 'microgliopathies', and those in which lysosomal or peroxisomal defects are considered to be the primary driver. The latter are suspected to have notable microglia involvement, as some affected individuals benefit from microglia-replenishing therapy. Based on overlapping pathology, we discuss multiple ways through which aberrant microglia could lead to white matter defects and brain dysfunction. We propose that the study of leukodystrophies, and their extensively multicellular pathology, will benefit from complementing analyses of human patient material with the examination of cellular dynamics in vivo using animal models, such as zebrafish. Together, this will yield important insight into the cell biological mechanisms of microglial impact in the central nervous system, particularly in the development and maintenance of myelin, that will facilitate the development of new, and refinement of existing, therapeutic options for a range of brain diseases.
Collapse
Affiliation(s)
| | | | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
25
|
The neuroprotective role of morroniside against spinal cord injury in female rats. Neurochem Int 2021; 148:105105. [PMID: 34147513 DOI: 10.1016/j.neuint.2021.105105] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 11/20/2022]
Abstract
Spinal cord injury (SCI) is a disabling condition that often leads to permanent neurological deficits without an effective treatment. Reactive oxygen species (ROS) produced during oxidative stress play a vital role in the pathogenesis following SCI. The antioxidant morroniside is the main active component of the Chinese medicine Cornus officinalis. In recent years, it has been reported that morroniside has therapeutic effects on damage to multiple organs mediated by oxidative damage, but the effect of morroniside on SCI has not been reported. The purpose of this study was therefore to assess the therapeutic effect of morroniside on SCI, and to identify its underlying mechanism by direct intragastric administration immediately after SCI. Our study showed that morroniside treatment improved the functional recovery of rats following SCI. This behavioral improvement was associated with the higher survival in neurons and oligodendrocytes following SCI, which increased the capacity of injured spinal cord (SC) to form myelin and repair tissue, eventually contributing to improved neurological outcome. Furthermore, our study found that oxygen free radicals increased and antioxidant enzyme activity decreased in the injured SC. Interestingly, morroniside treatment decreased oxygen free radical levels and increased antioxidant enzyme activities. Together, our results suggested that morroniside may be an effective treatment for improving outcomes following SCI, and that its antioxidant activity may be one of the mechanisms by which morroniside exerts neuroprotective effects on SCI.
Collapse
|
26
|
Kiper K, Freeman JL. Use of Zebrafish Genetic Models to Study Etiology of the Amyloid-Beta and Neurofibrillary Tangle Pathways in Alzheimer's Disease. Curr Neuropharmacol 2021; 20:524-539. [PMID: 34030617 DOI: 10.2174/1570159x19666210524155944] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/09/2021] [Accepted: 05/16/2021] [Indexed: 11/22/2022] Open
Abstract
The prevalence of neurodegenerative diseases is increasing globally, with an imperative need to identify and expand the availability of pharmaceutical treatment strategies. Alzheimer's disease is the most common neurodegenerative disease for which there is no cure or has limited treatments. Rodent models are primarily used in Alzheimer's disease research to investigate causes, pathology, molecular mechanisms, and pharmaceutical therapies. However, there is a lack of a comprehensive understanding of Alzheimer's disease causes, pathogenesis, and optimal treatments due in part to some limitations of using rodents, including higher economic cost, which can influence sample size and ultimately statistical power. It is necessary to expand our animal model toolbox to provide alternative strategies in Alzheimer's disease research. The zebrafish application in neurodegenerative disease research and neuropharmacology is greatly expanding due to several vital strengths spanning lower economic costs, the smaller size of the organism, a sequenced characterized genome, and well described anatomical structures. These characteristics are coupled to the conserved molecular function and disease pathways in humans. The existence of orthologs for genes associated with Alzheimer's disease in zebrafish is also confirmed. While wild-type zebrafish appear to lack some of the neuropathological features of Alzheimer's disease, the advent of genetic editing technologies has expanded evaluation of the amyloid and neurofibrillary tangle hypotheses using the zebrafish and exploration of pharmaceutical molecular targets. An overview of how genetic editing technologies are being used with the zebrafish to create models to investigate the causes, pathology, molecular mechanisms, and pharmaceutical targets of Alzheimer's disease is detailed.
Collapse
Affiliation(s)
- Keturah Kiper
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, Indiana, United States
| |
Collapse
|
27
|
MRI- and histologically derived neuroanatomical atlas of the Ambystoma mexicanum (axolotl). Sci Rep 2021; 11:9850. [PMID: 33972650 PMCID: PMC8110773 DOI: 10.1038/s41598-021-89357-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 04/12/2021] [Indexed: 02/03/2023] Open
Abstract
Amphibians are an important vertebrate model system to understand anatomy, genetics and physiology. Importantly, the brain and spinal cord of adult urodels (salamanders) have an incredible regeneration capacity, contrary to anurans (frogs) and the rest of adult vertebrates. Among these amphibians, the axolotl (Ambystoma mexicanum) has gained most attention because of the surge in the understanding of central nervous system (CNS) regeneration and the recent sequencing of its whole genome. However, a complete comprehension of the brain anatomy is not available. In the present study we created a magnetic resonance imaging (MRI) atlas of the in vivo neuroanatomy of the juvenile axolotl brain. This is the first MRI atlas for this species and includes three levels: (1) 82 regions of interest (ROIs) and a version with 64 ROIs; (2) a division of the brain according to the embryological origin of the neural tube, and (3) left and right hemispheres. Additionally, we localized the myelin rich regions of the juvenile brain. The atlas, the template that the atlas was derived from, and a masking file, can be found on Zenodo at https://doi.org/10.5281/zenodo.4595016 . This MRI brain atlas aims to be an important tool for future research of the axolotl brain and that of other amphibians.
Collapse
|
28
|
Hering I, Eilebrecht E, Parnham MJ, Weiler M, Günday-Türeli N, Türeli AE, Modh H, Heng PWS, Böhmer W, Schäfers C, Fenske M, Wacker MG. Microparticle formulations alter the toxicity of fenofibrate to the zebrafish Danio rerio embryo. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 234:105798. [PMID: 33799113 DOI: 10.1016/j.aquatox.2021.105798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 06/12/2023]
Abstract
A wide variety of active pharmaceutical ingredients are released into the environment and pose a threat to aquatic organisms. Drug products using micro- and nanoparticle technology can lower these emissions into the environment by their increased bioavailability to the human patients. However, due to this enhanced efficacy, micro- and nanoscale drug delivery systems can potentially display an even higher toxicity, and thus also pose a risk to non-target organisms. Fenofibrate is a lipid-regulating agent and exhibits species-related hazards in fish. The ecotoxic effects of a fenofibrate formulation embedded into a hydroxypropyl methylcellulose microparticle matrix, as well as those of the excipients used in the formulation process, were evaluated. To compare the effects of fenofibrate without a formulation, fenofibrate was dispersed in diluted ISO water alone or dissolved in the solvent DMF and then added to diluted ISO water. The effects of these various treatments were assessed using the fish embryo toxicity test, acridine orange staining and gene expression analysis assessed by quantitative RT polymerase chain reaction. Exposure concentrations were assessed by chemical analysis. The effect threshold concentrations of fenofibrate microparticle precipitates were higher compared to the formulation. Fenofibrate dispersed in 20%-ISO-water displayed the lowest toxicity. For the fenofibrate formulation as well as for fenofibrate added as a DMF solution, greater ecotoxic effects were observed in the zebrafish embryos. The chemical analysis of the solutions revealed that more fenofibrate was present in the samples with the fenofibrate formulation as well as fenofibrate added as a DMF solution compared to fenofibrate dispersed in diluted ISO water. This could explain the higher ecotoxicity. The toxic effects on the zebrafish embryo thus suggested that the formulation as well as the solvent increased the bioavailability of fenofibrate.
Collapse
Affiliation(s)
- Indra Hering
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596, Frankfurt/Main, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Auf dem Aberg 1, 57392, Schmallenberg, Germany; Goethe University Frankfurt am Main, Department Aquatic Ecotoxicology, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany.
| | - Elke Eilebrecht
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Auf dem Aberg 1, 57392, Schmallenberg, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596, Frankfurt/Main, Germany
| | - Marc Weiler
- MyBiotech GmbH, Industriestraße 1B, 66802, Überherrn, Germany
| | | | | | - Harshvardhan Modh
- National University of Singapore, Department of Pharmacy, Faculty of Science, Wet Science Building (S9), 5 Science Drive 2, 117546, Singapore, Singapore
| | - Paul W S Heng
- National University of Singapore, GEA-NUS Pharmaceutical Processing Research Laboratory, Department of Pharmacy, Faculty of Science, 18 Science Drive 4, 117543, Singapore, Singapore
| | - Walter Böhmer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Auf dem Aberg 1, 57392, Schmallenberg, Germany
| | - Christoph Schäfers
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Auf dem Aberg 1, 57392, Schmallenberg, Germany
| | - Martina Fenske
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Theodor-Stern-Kai 7, 60596, Frankfurt/Main, Germany.
| | - Matthias G Wacker
- National University of Singapore, Department of Pharmacy, Faculty of Science, Wet Science Building (S9), 5 Science Drive 2, 117546, Singapore, Singapore
| |
Collapse
|
29
|
Möbius W, Hümmert S, Ruhwedel T, Kuzirian A, Gould R. New Species Can Broaden Myelin Research: Suitability of Little Skate, Leucoraja erinacea. Life (Basel) 2021; 11:136. [PMID: 33670172 PMCID: PMC7916940 DOI: 10.3390/life11020136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/02/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023] Open
Abstract
Although myelinated nervous systems are shared among 60,000 jawed vertebrates, studies aimed at understanding myelination have focused more and more on mice and zebrafish. To obtain a broader understanding of the myelination process, we examined the little skate, Leucoraja erinacea. The reasons behind initiating studies at this time include: the desire to study a species belonging to an out group of other jawed vertebrates; using a species with embryos accessible throughout development; the availability of genome sequences; and the likelihood that mammalian antibodies recognize homologs in the chosen species. We report that the morphological features of myelination in a skate hatchling, a stage that supports complex behavioral repertoires needed for survival, are highly similar in terms of: appearances of myelinating oligodendrocytes (CNS) and Schwann cells (PNS); the way their levels of myelination conform to axon caliber; and their identity in terms of nodal and paranodal specializations. These features provide a core for further studies to determine: axon-myelinating cell communication; the structures of the proteins and lipids upon which myelinated fibers are formed; the pathways used to transport these molecules to sites of myelin assembly and maintenance; and the gene regulatory networks that control their expressions.
Collapse
Affiliation(s)
- Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, 37075 Göttingen, Germany; (W.M.); (S.H.); (T.R.)
- Cluster of Excellence Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, 37073 Göttingen, Germany
| | - Sophie Hümmert
- Electron Microscopy Core Unit, Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, 37075 Göttingen, Germany; (W.M.); (S.H.); (T.R.)
| | - Torben Ruhwedel
- Electron Microscopy Core Unit, Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, 37075 Göttingen, Germany; (W.M.); (S.H.); (T.R.)
| | - Alan Kuzirian
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, Woods Hole, MA 02540, USA;
| | - Robert Gould
- Whitman Science Center, Marin Biological Laboratory, Woods Hole, MA 02540, USA
| |
Collapse
|
30
|
Cao P, Zhang H, Meng H, Cheng Y, Xu H, Zang S, Li Z, Cui J, Li Y. Celecoxib Exerts a Therapeutic Effect Against Demyelination by Improving the Immune and Inflammatory Microenvironments. J Inflamm Res 2020; 13:1043-1055. [PMID: 33293848 PMCID: PMC7718997 DOI: 10.2147/jir.s282128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/11/2020] [Indexed: 12/29/2022] Open
Abstract
Background The myelin sheath can be damaged by genetic and/or environmental factors, leading to demyelinating diseases, for which effective treatments are lacking. Recently, cyclooxygenase-2 (COX-2) overexpression was detected in demyelinating lesions both in patients and animal models, opening an avenue for promoting endogenous remyelination. The aim of this study was to investigate the therapeutic effect of celecoxib, a selective COX-2 inhibitor, against demyelination in a zebrafish model. Methods The biotoxicity of celecoxib was evaluated on zebrafish embryos. Metronidazole was used to deplete the oligodendrocytes in Tg (mbp:nfsB-egfp) transgenic fish. Celecoxib was then administered both in larvae and adults. The regeneration of the myelin sheath and the underlying mechanisms were explored by immunohistochemistry, flow cytometry, Western blot analysis, quantitative real-time polymerase chain reaction, and behavioral test. Results Celecoxib had low in vivo toxicity. A stable and practical demyelination model was established by metronidazole induction. Following celecoxib treatment, the number of oligodendrocytes was increased significantly and the concentric structure of the myelin sheath reappeared. The locomotor ability was notably improved and was close to its physiological levels. The expression of arg1, mrc1, il-10, and il-4 was upregulated, while that of il-1β, il-12, tnf-α, il-6, caspase-3 and caspase-7 was downregulated. Conclusion Inhibition of COX-2 contributed to the transformation of microglia/macrophages from the M1 to the M2 phenotype, improved the inflammatory microenvironment, and suppressed caspase-dependent apoptosis, thus exerting a therapeutic effect against demyelination.
Collapse
Affiliation(s)
- Peipei Cao
- Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Hao Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Huiling Meng
- Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Yajia Cheng
- Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Haiqi Xu
- Faculty of Life Science, University of Liverpool, Liverpool, UK
| | - Siwen Zang
- Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Jianlin Cui
- Nankai University School of Medicine, Tianjin, People's Republic of China.,Medical International Collaborative Innovation Center, Nankai University School of Medicine, Tianjin, People's Republic of China
| | - Yuhao Li
- Nankai University School of Medicine, Tianjin, People's Republic of China.,Department of Pathology, Nankai University School of Medicine, Tianjin, People's Republic of China
| |
Collapse
|
31
|
Molecular Mechanisms of Central Nervous System Axonal Regeneration and Remyelination: A Review. Int J Mol Sci 2020; 21:ijms21218116. [PMID: 33143194 PMCID: PMC7662268 DOI: 10.3390/ijms21218116] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Central nervous system (CNS) injury, including stroke, spinal cord injury, and traumatic brain injury, causes severe neurological symptoms such as sensory and motor deficits. Currently, there is no effective therapeutic method to restore neurological function because the adult CNS has limited capacity to regenerate after injury. Many efforts have been made to understand the molecular and cellular mechanisms underlying CNS regeneration and to establish novel therapeutic methods based on these mechanisms, with a variety of strategies including cell transplantation, modulation of cell intrinsic molecular mechanisms, and therapeutic targeting of the pathological nature of the extracellular environment in CNS injury. In this review, we will focus on the mechanisms that regulate CNS regeneration, highlighting the history, recent efforts, and questions left unanswered in this field.
Collapse
|
32
|
Zabegalov KN, Wang D, Yang L, Wang J, Hu G, Serikuly N, Alpyshov ET, Khatsko SL, Zhdanov A, Demin KA, Galstyan DS, Volgin AD, de Abreu MS, Strekalova T, Song C, Amstislavskaya TG, Sysoev Y, Musienko PE, Kalueff AV. Decoding the role of zebrafish neuroglia in CNS disease modeling. Brain Res Bull 2020; 166:44-53. [PMID: 33027679 DOI: 10.1016/j.brainresbull.2020.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/14/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
Neuroglia, including microglia and astrocytes, is a critical component of the central nervous system (CNS) that interacts with neurons to modulate brain activity, development, metabolism and signaling pathways. Thus, a better understanding of the role of neuroglia in the brain is critical. Complementing clinical and rodent data, the zebrafish (Danio rerio) is rapidly becoming an important model organism to probe the role of neuroglia in brain disorders. With high genetic and physiological similarity to humans and rodents, zebrafish possess some common (shared), as well as some specific molecular biomarkers and features of neuroglia development and functioning. Studying these common and zebrafish-specific aspects of neuroglia may generate important insights into key brain mechanisms, including neurodevelopmental, neurodegenerative, neuroregenerative and neurological processes. Here, we discuss the biology of neuroglia in humans, rodents and fish, its role in various CNS functions, and further directions of translational research into the role of neuroglia in CNS disorders using zebrafish models.
Collapse
Affiliation(s)
- Konstantin N Zabegalov
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia
| | - Dongmei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - LongEn Yang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Jingtao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Guojun Hu
- School of Pharmacy, Southwest University, Chongqing, China
| | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | | | | | | | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Andrey D Volgin
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil; Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia.
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Division of Molecular Psychiatry, Centre of Mental Health, University of Würzburg, Würzburg, Germany
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Guangdong Ocean University, Zhanjiang, China; Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Shenzhen, China
| | - Tamara G Amstislavskaya
- Scientific Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Zelman Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Yury Sysoev
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Department of Pharmacology and Clinical Pharmacology, St. Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | - Pavel E Musienko
- Laboratory of Neuroprosthetics, Institute of Translational Biomedicine, Petersburg State University, St. Petersburg, Russia; Institute of Phthisiopulmonology, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
33
|
Shen C, Zhou Y, Tang C, He C, Zuo Z. Developmental exposure to mepanipyrim induces locomotor hyperactivity in zebrafish (Danio rerio) larvae. CHEMOSPHERE 2020; 256:127106. [PMID: 32447115 DOI: 10.1016/j.chemosphere.2020.127106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 06/11/2023]
Abstract
Mepanipyrim is a widely used fungicide, and residues of mepanipyrim are frequently detected in commodities. However, the neurotoxicity and underlying mechanisms of mepanipyrim are still insufficiently understood. In this study, zebrafish embryos at 0.5-1.0 post-fertilization hours (hpf) were exposed to 0.1, 1, 10 and 100 μg/L mepanipyrim for 7 days. Our results showed that mepanipyrim could cause the locomotor hyperactivity and increase the concentration of γ-amino butyric acid (GABA) and the Na+/K+- and Ca2+-ATPase activities in zebrafish larvae. We have conducted the RNA-sequence and RT-qPCR to analyze the gene expressions. The mRNA expression levels of calcium/sodium ion conduction associated genes were observably up-regulated, demonstrating that mepanipyrim could enhance the cell energy metabolism, the synaptic transmission and skeletal muscle contraction, which were consistent with the locomotor hyperactivity. Meanwhile, exposure to mepanipyrim could significantly change the gene expression levels of gad1, bdnf, nlgn1, and type A and B GABA receptors in zebrafish larvae. This is the first study focusing on the underlying mechanisms of the neurotoxic effects that are induced by mepanipyrim.
Collapse
Affiliation(s)
- Chao Shen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yixi Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chen Tang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
34
|
Zhang T, Peterson RT. Modeling Lysosomal Storage Diseases in the Zebrafish. Front Mol Biosci 2020; 7:82. [PMID: 32435656 PMCID: PMC7218095 DOI: 10.3389/fmolb.2020.00082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Lysosomal storage diseases (LSDs) are a family of 70 metabolic disorders characterized by mutations in lysosomal proteins that lead to storage material accumulation, multiple-organ pathologies that often involve neurodegeneration, and early mortality in a significant number of patients. Along with the necessity for more effective therapies, there exists an unmet need for further understanding of disease etiology, which could uncover novel pathways and drug targets. Over the past few decades, the growth in knowledge of disease-associated pathways has been facilitated by studies in model organisms, as advancements in mutagenesis techniques markedly improved the efficiency of model generation in mammalian and non-mammalian systems. In this review we highlight non-mammalian models of LSDs, focusing specifically on the zebrafish, a vertebrate model organism that shares remarkable genetic and metabolic similarities with mammals while also conferring unique advantages such as optical transparency and amenability toward high-throughput applications. We examine published zebrafish LSD models and their reported phenotypes, address organism-specific advantages and limitations, and discuss recent technological innovations that could provide potential solutions.
Collapse
Affiliation(s)
- T Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States
| | - R T Peterson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
35
|
Ding S, Gu Y, Cai Y, Cai M, Yang T, Bao S, Shen W, Ni X, Chen G, Xing L. Integrative systems and functional analyses reveal a role of dopaminergic signaling in myelin pathogenesis. J Transl Med 2020; 18:109. [PMID: 32122379 PMCID: PMC7053059 DOI: 10.1186/s12967-020-02276-1] [Citation(s) in RCA: 9] [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: 11/11/2019] [Accepted: 02/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Myelin sheaths surrounding axons are critical for electrical signal transmission in the central nervous system (CNS). Diseases with myelin defects such as multiple sclerosis (MS) are devastating neurological conditions for which few effective treatments are available. Dysfunction of the dopaminergic system has been observed in multiple neurological disorders. Its role in myelin pathogenesis, however, is unclear. METHODS This work used a combination of literature curation, bioinformatics, pharmacological and genetic manipulation, as well as confocal imaging techniques. Literature search was used to establish a complete set of genes which is associated with MS in humans. Bioinformatics analyses include pathway enrichment and crosstalk analyses with human genetic association studies as well as gene set enrichment and causal relationship analyses with transcriptome data. Pharmacological and CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) genetic manipulation were applied to inhibit the dopaminergic signaling in zebrafish. Imaging techniques were used to visualize myelin formation in vivo. RESULTS Systematic analysis of human genetic association studies revealed that the dopaminergic synapse signaling pathway is enriched in candidate gene sets. Transcriptome analysis confirmed that expression of multiple dopaminergic gene sets was significantly altered in patients with MS. Pathway crosstalk analysis and gene set causal relationship analysis reveal that the dopaminergic synapse signaling pathway interacts with or is associated with other critical pathways involved in MS. We also found that disruption of the dopaminergic system leads to myelin deficiency in zebrafish. CONCLUSIONS Dopaminergic signaling may be involved in myelin pathogenesis. This study may offer a novel molecular mechanism of demyelination in the nervous system.
Collapse
Affiliation(s)
- Sujun Ding
- School of Medicine, Nantong University, Nantong, China
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yunyun Cai
- Department of Physiology, School of medicine, Nantong University, Nantong, China
| | - Meijuan Cai
- Department of Clinical Laboratory, Qilu Hospital of Shandong university, Shandong, China
| | - Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shuangxi Bao
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Weixing Shen
- Department of Physiology, School of medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xuejun Ni
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong, China
| | - Gang Chen
- School of Medicine, Nantong University, Nantong, China
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Lingyan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
36
|
Demin KA, Lakstygal AM, Chernysh MV, Krotova NA, Taranov AS, Ilyin NP, Seredinskaya MV, Tagawa N, Savva AK, Mor MS, Vasyutina ML, Efimova EV, Kolesnikova TO, Gainetdinov RR, Strekalova T, Amstislavskaya TG, de Abreu MS, Kalueff AV. The zebrafish tail immobilization (ZTI) test as a new tool to assess stress-related behavior and a potential screen for drugs affecting despair-like states. J Neurosci Methods 2020; 337:108637. [PMID: 32081675 DOI: 10.1016/j.jneumeth.2020.108637] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Affective disorders, especially depression and anxiety, are highly prevalent, debilitating mental illnesses. Animal experimental models are a valuable tool in translational affective neuroscience research. A hallmark phenotype of clinical and experimental depression, the learned helplessness, has become a key target for 'behavioral despair'-based animal models of depression. The zebrafish (Danio rerio) has recently emerged as a promising novel organism for affective disease modeling and CNS drug screening. Despite being widely used to assess stress and anxiety-like behaviors, there are presently no clear-cut despair-like models in zebrafish. NEW METHOD Here, we introduce a novel behavioral paradigm, the zebrafish tail immobilization (ZTI) test, as a potential tool to assess zebrafish despair-like behavior. Conceptually similar to rodent 'despair' models, the ZTI protocol involves immobilizing the caudal half of the fish body for 5 min, leaving the cranial part to move freely, suspended vertically in a small beaker with water. RESULTS To validate this model, we used exposure to low-voltage electric shock, alarm pheromone, selected antidepressants (sertraline and amitriptyline) and an anxiolytic drug benzodiazepine (phenazepam), assessing the number of mobility episodes, time spent 'moving', total distance moved and other activity measures of the cranial part of the body, using video-tracking. Both electric shock and alarm pheromone decreased zebrafish activity in this test, antidepressants increased it, and phenazepam was inactive. Furthermore, a 5-min ZTI exposure increased serotonin turnover, elevating the 5-hydroxyindoleacetic acid/serotonin ratio in zebrafish brain, while electric shock prior to ZTI elevated both this and the 3,4-dihydroxyphenylacetic acid/dopamine ratios. In contrast, preexposure to antidepressants sertraline and amitriptyline lowered both ratios, compared to the ZTI test-exposed fish. COMPARISON WITH EXISTINGMETHOD(S) The ZTI test is the first despair-like experimental model in zebrafish. CONCLUSIONS Collectively, this study suggests the ZTI test as a potentially useful protocol to assess stress-/despair-related behaviors, potentially relevant to CNS drug screening and behavioral phenotyping of zebrafish.
Collapse
Affiliation(s)
- Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
| | - Anton M Lakstygal
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Maria V Chernysh
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Natalia A Krotova
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Aleksandr S Taranov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Nikita P Ilyin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Maria V Seredinskaya
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Natsuki Tagawa
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Anna K Savva
- Laboratory of Insect Biopharmacology and Immunology, Faculty of Biology, St. Petersburg State University, St. Petersburg, Russia
| | - Mikael S Mor
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Marina L Vasyutina
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Evgeniya V Efimova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatyana O Kolesnikova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatyana Strekalova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Maastricht University, Maastricht, The Netherlands; Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| | | | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
37
|
Torre-Fuentes L, Moreno-Jiménez L, Pytel V, Matías-Guiu J, Gómez-Pinedo U, Matías-Guiu J. Experimental models of demyelination and remyelination. NEUROLOGÍA (ENGLISH EDITION) 2020. [PMCID: PMC7148713 DOI: 10.1016/j.nrleng.2019.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
38
|
Vancamp P, Demeneix BA, Remaud S. Monocarboxylate Transporter 8 Deficiency: Delayed or Permanent Hypomyelination? Front Endocrinol (Lausanne) 2020; 11:283. [PMID: 32477268 PMCID: PMC7237703 DOI: 10.3389/fendo.2020.00283] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022] Open
Abstract
Monocarboxylate transporter 8 (MCT8) deficiency or the Allan-Herndon-Dudley Syndrome (AHDS) is an X-linked psychomotor disability syndrome with around 320 clinical cases described worldwide. SLC16A2 gene mutations, encoding the thyroid hormone (TH) transporter MCT8, result in intellectual disability due to impaired TH uptake in the developing brain. MCT8 deficiency is a multi-organ affecting disease with a predominant neuronal cell-based pathology, with the glial component inadequately investigated. However, deficiency in myelin, a key component of white matter (WM) enabling fast nerve conduction, is a TH-dependent hallmark of the disease. Nevertheless, analysis of the myelin status in AHDS patients has led to conflicting interpretations. The majority of individual case studies reported delayed myelination, that was restored later in life. In contrast, post-mortem studies and high-resolution MRIs detected WM (micro-) abnormalities throughout adolescence, suggesting permanent hypomyelination. Thus, interpretations vary depending on methodology to investigate WM microstructure. Further, it is unknown whether the mutation within the MCT8 is linked to the severity of the myelin deficiency. Consequently, terminology is inconsistent among reports, and AHDS is occasionally misdiagnosed as another WM disorder. The evolutionary conserved TH signaling pathway that promotes the generation of myelinating oligodendrocytes enabled deciphering how the lack of MCT8 might affect myelinogenesis. Linking patient findings on myelination to those obtained from models of MCT8 deficiency revealed underlying pathophysiological mechanisms, but knowledge gaps remain, notably how myelination progresses both spatially and temporally in MCT8 deficiency. This limits predicting how myelin integrity might benefit therapeutically, and when to initiate. A recurrent observation in clinical trials is the absence of neurological improvement. Testing MCT8-independent thyromimetics in models, and evaluating treatments used in other demyelinating diseases, despite different etiologies, is crucial to propose new therapeutic strategies combatting this devastating disease.
Collapse
Affiliation(s)
- Pieter Vancamp
- UMR 7221 Molecular Physiology and Adaptation, Centre National de le Recherche Scientifique-Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Molecular Physiology and Adaptation, Centre National de le Recherche Scientifique-Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Molecular Physiology and Adaptation, Centre National de le Recherche Scientifique-Muséum National d'Histoire Naturelle, Paris, France
| |
Collapse
|
39
|
Nagarajan B, Harder A, Japp A, Häberlein F, Mingardo E, Kleinert H, Yilmaz Ö, Zoons A, Rau B, Christ A, Kubitscheck U, Eiberger B, Sandhoff R, Eckhardt M, Hartmann D, Odermatt B. CNS myelin protein 36K regulates oligodendrocyte differentiation through Notch. Glia 2019; 68:509-527. [PMID: 31702067 DOI: 10.1002/glia.23732] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 12/12/2022]
Abstract
In contrast to humans and other mammals, zebrafish can successfully regenerate and remyelinate central nervous system (CNS) axons following injury. In addition to common myelin proteins found in mammalian myelin, 36K protein is a major component of teleost fish CNS myelin. Although 36K is one of the most abundant proteins in zebrafish brain, its function remains unknown. Here we investigate the function of 36K using translation-blocking Morpholinos. Morphant larvae showed fewer dorsally migrated oligodendrocyte precursor cells as well as upregulation of Notch ligand. A gamma secretase inhibitor, which prevents activation of Notch, could rescue oligodendrocyte precursor cell numbers in 36K morphants, suggesting that 36K regulates initial myelination through inhibition of Notch signaling. Since 36K like other short chain dehydrogenases might act on lipids, we performed thin layer chromatography and mass spectrometry of lipids and found changes in lipid composition in 36K morphant larvae. Altogether, we suggest that during early development 36K regulates membrane lipid composition, thereby altering the amount of transmembrane Notch ligands and the efficiency of intramembrane gamma secretase processing of Notch and thereby influencing oligodendrocyte precursor cell differentiation and further myelination. Further studies on the role of 36K short chain dehydrogenase in oligodendrocyte precursor cell differentiation during remyelination might open up new strategies for remyelination therapies in human patients.
Collapse
Affiliation(s)
- Bhuvaneswari Nagarajan
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Alexander Harder
- Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | - Anna Japp
- Institute of Neuropathology, University Clinics, University of Bonn, Bonn, Germany
| | - Felix Häberlein
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany.,Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Enrico Mingardo
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Henning Kleinert
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Öznur Yilmaz
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Angelika Zoons
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Birgit Rau
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Andrea Christ
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Ulrich Kubitscheck
- Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | - Britta Eiberger
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Centre, Heidelberg, Germany
| | - Matthias Eckhardt
- Institute of Biochemistry and Molecular Biology, University Clinics, University of Bonn, Bonn, Germany
| | - Dieter Hartmann
- Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| | - Benjamin Odermatt
- Institute of Anatomy, Division of Anatomy and Cell Biology, University Clinics, University of Bonn, Bonn, Germany.,Institute of Anatomy, Division of Neuroanatomy, University Clinics, University of Bonn, Bonn, Germany
| |
Collapse
|
40
|
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: 2.3] [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
|
41
|
Zhu XY, Guo SY, Xia B, Li CQ, Wang L, Wang YH. Development of zebrafish demyelination model for evaluation of remyelination compounds and RORγt inhibitors. J Pharmacol Toxicol Methods 2019; 98:106585. [DOI: 10.1016/j.vascn.2019.106585] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 05/08/2019] [Accepted: 05/15/2019] [Indexed: 11/25/2022]
|
42
|
Adler D, Linden JR, Shetty SV, Ma Y, Bokori-Brown M, Titball RW, Vartanian T. Clostridium perfringens Epsilon Toxin Compromises the Blood-Brain Barrier in a Humanized Zebrafish Model. iScience 2019; 15:39-54. [PMID: 31030181 PMCID: PMC6487375 DOI: 10.1016/j.isci.2019.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/29/2018] [Accepted: 04/08/2019] [Indexed: 12/22/2022] Open
Abstract
Clostridium perfringens epsilon toxin (ETX) is hypothesized to mediate blood-brain barrier (BBB) permeability by binding to the myelin and lymphocyte protein (MAL) on the luminal surface of endothelial cells (ECs). However, the kinetics of this interaction and a general understanding of ETX's behavior in a live organism have yet to be appreciated. Here we investigate ETX binding and BBB breakdown in living Danio rerio (zebrafish). Wild-type zebrafish ECs do not bind ETX. When zebrafish ECs are engineered to express human MAL (hMAL), proETX binding occurs in a time-dependent manner. Injection of activated toxin in hMAL zebrafish initiates BBB leakage, hMAL downregulation, blood vessel stenosis, perivascular edema, and blood stasis. We propose a kinetic model of MAL-dependent ETX binding and neurovascular pathology. By generating a humanized zebrafish BBB model, this study contributes to our understanding of ETX-induced BBB permeability and strengthens the proposal that MAL is the ETX receptor.
Collapse
Affiliation(s)
- Drew Adler
- Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; Department of Neurobiology and Behavior, Cornell University, Ithaca, NY 14850, USA
| | - Jennifer R Linden
- Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Samantha V Shetty
- Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Yinghua Ma
- Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | | | - Richard W Titball
- Department of Biosciences, University of Exeter, Exeter, Devon EX4 4SB, UK
| | - Timothy Vartanian
- Brain and Mind Research Institute, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
43
|
Hill RA, Grutzendler J. Uncovering the biology of myelin with optical imaging of the live brain. Glia 2019; 67:2008-2019. [PMID: 31033062 DOI: 10.1002/glia.23635] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/26/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Myelin has traditionally been considered a static structure that is produced and assembled during early developmental stages. While this characterization is accurate in some contexts, recent studies have revealed that oligodendrocyte generation and patterns of myelination are dynamic and potentially modifiable throughout life. Unique structural and biochemical properties of the myelin sheath provide opportunities for the development and implementation of multimodal label-free and fluorescence optical imaging approaches. When combined with genetically encoded fluorescent tags targeted to distinct cells and subcellular structures, these techniques offer a powerful methodological toolbox for uncovering mechanisms of myelin generation and plasticity in the live brain. Here, we discuss recent advances in these approaches that have allowed the discovery of several forms of myelin plasticity in developing and adult nervous systems. Using these techniques, long-standing questions related to myelin generation, remodeling, and degeneration can now be addressed.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire
| | - Jaime Grutzendler
- Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
44
|
Preston MA, Finseth LT, Bourne JN, Macklin WB. A novel myelin protein zero transgenic zebrafish designed for rapid readout of in vivo myelination. Glia 2019; 67:650-667. [PMID: 30623975 PMCID: PMC6555554 DOI: 10.1002/glia.23559] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 12/14/2022]
Abstract
Demyelination occurs following many neurological insults, most notably in multiple sclerosis (MS). Therapeutics that promote remyelination could slow the neurological decline associated with chronic demyelination; however, in vivo testing of candidate small molecule drugs and signaling cascades known to impact myelination is expensive and labor intensive. Here, we describe the development of a novel zebrafish line which uses the putative promoter of Myelin Protein Zero (mpz), a major structural protein in myelin, to drive expression of Enhanced Green Fluorescent Protein (mEGFP) specifically in the processes and nascent internodes of myelinating glia. We observe that changes in fluorescence intensity in Tg(mpz:mEGFP) larvae are a reliable surrogate for changes in myelin membrane production per se in live larvae following bath application of drugs. These changes in fluorescence are strongly predictive of changes in myelin-specific mRNAs [mpz, 36K and myelin basic protein (mbp)] and protein production (Mbp). Finally, we observe that certain drugs alter nascent internode number and length, impacting the overall amount of myelin membrane synthesized and a number of axons myelinated without significantly changing the number of myelinating oligodendrocytes. These studies demonstrate that the Tg(mpz:mEGFP) reporter line responds effectively to positive and negative small molecule regulators of myelination, and could be useful for identifying candidate drugs that specifically target myelin membrane production in vivo. Combined with high throughput cell-based screening of large chemical libraries and automated imaging systems, this transgenic line is useful for rapid large scale whole animal screening to identify novel myelinating small molecule compounds in vivo.
Collapse
Affiliation(s)
- Marnie A Preston
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Lisbet T Finseth
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jennifer N Bourne
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
45
|
Lazcano I, Rodríguez-Ortiz R, Villalobos P, Martínez-Torres A, Solís-Saínz JC, Orozco A. Knock-Down of Specific Thyroid Hormone Receptor Isoforms Impairs Body Plan Development in Zebrafish. Front Endocrinol (Lausanne) 2019; 10:156. [PMID: 30930855 PMCID: PMC6427925 DOI: 10.3389/fendo.2019.00156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
The role of thyroid hormones (THs) in development has been extensively studied, however, the specific molecular mechanisms involved are far from being clear. THs act by binding to TH nuclear receptors (TR) that act as ligand-dependent transcription factors to regulate TH-dependent gene expression. Like vertebrates, zebrafish express different isoforms of functional Tr alpha and beta, some of which can bind alternative ligands like 3,5-T2. In this study, we first analyzed the effects of exogenous T3 and 3,5-T2 exposure during embryogenesis. The percentage of affected embryos was similar to those vehicle-injected, suggesting that the early exposure to low TH levels is not sufficient to elicit effects upon the phenotype of the embryo. We then generated crispants for four isoforms of thr to learn more about the role of these receptors in early development. We found that crispant larvae from thraa and a newly identified l-thrb+, but not thrab and canonical thrb1 showed profound deleterious effects upon symmetry and laterality, suggesting early novel roles for these Tr isoforms in the body plan developmental program. Since critical events that determine cell fate start in the late gastrula, we tested if some genes that are expressed during early developmental stages could indeed be TH targets. We identify early development genes, like sox10 and eve, that were specifically over-expressed in thraa and l-thrb+ crispants, suggesting that these specific thr isoforms function as transcription repressors for these genes, while transcription of zic and ets appear to be thraa and l-thrb+-mediated, respectively. Overall, present results show that TH signaling participates in early zebrafish development and identify Tr isoform-specific mediated regulation of early gene expression.
Collapse
Affiliation(s)
- Iván Lazcano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- Departmento de Investigación Biomédica, Facultad de Medicina, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Roberto Rodríguez-Ortiz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- CONACYT – Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Patricia Villalobos
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Ataúlfo Martínez-Torres
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Juan Carlos Solís-Saínz
- Departmento de Investigación Biomédica, Facultad de Medicina, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Aurea Orozco
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- *Correspondence: Aurea Orozco
| |
Collapse
|
46
|
Manipulating Neuronal Activity in the Developing Zebrafish Spinal Cord to Investigate Adaptive Myelination. Methods Mol Biol 2019; 1936:211-225. [PMID: 30820901 DOI: 10.1007/978-1-4939-9072-6_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the central nervous system, oligodendrocyte-lineage cells and myelination can adapt to physiological brain activity. Since myelin can in turn regulate neuronal function, such "adaptive" myelination has been proposed as a form of nervous system plasticity, implicated in learning and cognition. The molecular and cellular mechanisms underlying adaptive myelination and its functional consequences remain to be fully defined, partly because it remains challenging to manipulate activity and monitor myelination over time in vivo at single-cell resolution, in a model that would also allow examination of the functional output of individual neurons and circuits. Here, we describe a workflow to manipulate neuronal activity and to assess oligodendrocyte-lineage cell dynamics and myelination in larval zebrafish, a vertebrate animal model that is ideal for live imaging and amenable to genetic discovery, and that has well-characterized neuronal circuits with myelinated axons.
Collapse
|
47
|
Treichel AJ, Hines JH. Development of an Embryonic Zebrafish Oligodendrocyte-Neuron Mixed Coculture System. Zebrafish 2018; 15:586-596. [PMID: 30300571 DOI: 10.1089/zeb.2018.1625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
During vertebrate neural development, oligodendrocytes insulate nerve axons with myelin sheaths. Zebrafish (Danio rerio) has emerged as a useful model organism for studying oligodendrocyte development. However, the absence of an in vitro culture system necessitates in vivo manipulations and analyses, which, in some instances, limits the questions that can be addressed. To fill this gap we developed a mixed coculture system for embryonic zebrafish neurons and oligodendrocyte-lineage cells. Cultures harvested from embryos ≥30 hours postfertilization (hpf) yielded oligodendrocyte progenitor cells (OPCs) positive for olig2 and sox10 transgenic reporters. Cultured OPCs exhibited dynamic, exploratory membrane processes, and cell morphologies resembled those established in vivo. Cells harvested from advanced stage embryos possessed more arborized processes than those from early stage embryos. Advanced stage (>60 hpf) embryo culture produced differentiated, mbp+ oligodendrocytes. Genetically tractable neuron subtypes extended neurites when harvested from embryos ≥19 hpf. Coculture produced juxtaposed oligodendrocytes and neurons, demonstrating the practical usefulness of this technique for future studies examining axon-oligodendrocyte interactions under defined conditions. We expect that zebrafish oligodendrocyte culture will complement existing in vivo strengths and may facilitate future studies elucidating the mechanisms of oligodendrocyte specification, proliferation, differentiation, motility, and axon-oligodendrocyte interactions that shape adult myelination patterns.
Collapse
Affiliation(s)
| | - Jacob H Hines
- Department of Biology, Winona State University , Winona, Minnesota
| |
Collapse
|
48
|
Vancamp P, Darras VM. From zebrafish to human: A comparative approach to elucidate the role of the thyroid hormone transporter MCT8 during brain development. Gen Comp Endocrinol 2018; 265:219-229. [PMID: 29183795 DOI: 10.1016/j.ygcen.2017.11.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/24/2017] [Accepted: 11/24/2017] [Indexed: 02/07/2023]
Abstract
Monocarboxylate transporter 8 (MCT8) facilitates transmembrane transport of thyroid hormones (THs) ensuring their action on gene expression during vertebrate neurodevelopment. A loss of MCT8 in humans results in severe psychomotor deficits associated with the Allan-Herndon-Dudley Syndrome (AHDS). However, where and when exactly a lack of MCT8 causes the neurological manifestations remains unclear because of the varying expression pattern of MCT8 between specific brain regions and cells. Here, we elaborate on the animal models that have been generated to elucidate the mechanisms underlying MCT8-deficient brain development. The absence of a clear neurological phenotype in Mct8 knockout mice made it clear that a single species would not suffice. The evolutionary conservation of TH action on neurodevelopment as well as the components regulating TH signalling however offers the opportunity to answer different aspects of MCT8 function in brain development using different vertebrate species. Moreover, the plethora of tools for genome editing available today facilitates gene silencing in these animals as well. Studies in the recently generated mct8-deficient zebrafish and Mct8/Oatp1c1 double knockout mice have put forward the current paradigm of impaired TH uptake at the level of the blood-brain barrier during peri- and postnatal development as being the main pathophysiological mechanism of AHDS. RNAi vector-based, cell-specific induction of MCT8 knockdown in the chicken embryo points to an additional function of MCT8 at the level of the neural progenitors during early brain development. Future studies including also additional in vivo models like Xenopus or in vitro approaches such as induced pluripotent stem cells will continue to help unravelling the exact role of MCT8 in developmental events. In the end, this multispecies approach will lead to a unifying thesis regarding the cellular and molecular mechanisms responsible for the neurological phenotype in AHDS patients.
Collapse
Affiliation(s)
- Pieter Vancamp
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000 Leuven, Belgium
| | - Veerle M Darras
- KU Leuven, Laboratory of Comparative Endocrinology, Department of Biology, B-3000 Leuven, Belgium.
| |
Collapse
|
49
|
Özdemir S, Altun S, Özkaraca M, Ghosi A, Toraman E, Arslan H. Cypermethrin, chlorpyrifos, deltamethrin, and imidacloprid exposure up-regulates the mRNA and protein levels of bdnf and c-fos in the brain of adult zebrafish (Danio rerio). CHEMOSPHERE 2018; 203:318-326. [PMID: 29626809 DOI: 10.1016/j.chemosphere.2018.03.190] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 06/08/2023]
Abstract
The aim of the present study is to investigate the toxicity effects of frequently used pesticides, involving cypermethrin, deltamethrin, chlorpyrifos and imidacloprid, on the expression of bdnf and c-fos genes in zebrafish brain tissues. Therefore, brain tissues exposed to intoxication was primarily analyzed by indirect immunofluorescence assay. Afterwards, the mRNA transcription levels of BNDF and c-fos genes and the protein levels were measured by qRT-PCR and Western blotting, respectively. The data of the immunofluorescence assay revealed intensive immunopositivity for bdnf and c-fos genes in the tissues exposed to pesticide intoxication in comparison to the control group (p<0.05). Moreover, the transcription levels of BNDF and c-fos genes, and protein levels were elevated following the intoxication (p<0.05, p<0.01, and p<0.001, respectively). These results showed that the exposure to the acute cypermethrin, deltamethrin, chlorpyrifos and imidacloprid intoxication disrupted the normal neuronal activity, resulting in neurotoxic effect, also DNA-binding Increasing c-fos activation, an oncoprotein from the family of the Nuclear Proteins, is also true of the knowledge that these chemicals are oncogenic in zebrafish brain tissues. Thus, the use of these pesticides poses a potential neuronal and oncogenic risk to the non-target organisms.
Collapse
Affiliation(s)
- Selçuk Özdemir
- Department of Genetics, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240, Erzurum, Turkey.
| | - Serdar Altun
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240, Erzurum, Turkey
| | - Mustafa Özkaraca
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Yakutiye, 25240, Erzurum, Turkey
| | - Atena Ghosi
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Yakutiye, 25240, Erzurum, Turkey
| | - Emine Toraman
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, Yakutiye, 25240, Erzurum, Turkey
| | - Harun Arslan
- Department of Basic Sciences, Faculty of Fisheries, Atatürk University, Yakutiye, 25240, Erzurum, Turkey
| |
Collapse
|
50
|
Abu-Rub M, Miller RH. Emerging Cellular and Molecular Strategies for Enhancing Central Nervous System (CNS) Remyelination. Brain Sci 2018; 8:brainsci8060111. [PMID: 29914096 PMCID: PMC6024921 DOI: 10.3390/brainsci8060111] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
Myelination is critical for the normal functioning of the central nervous system (CNS) in vertebrates. Conditions in which the development of myelin is perturbed result in severely compromised individuals often with shorter lifespans, while loss of myelin in the adult results in a variety of functional deficits. Although some form of spontaneous remyelination often takes place, the repair process as a whole often fails. Several lines of evidence suggest it is feasible to develop strategies that enhance the capacity of the CNS to undergo remyelination and potentially reverse functional deficits. Such strategies include cellular therapies using either neural or mesenchymal stem cells as well as molecular regulators of oligodendrocyte development and differentiation. Given the prevalence of demyelinating diseases and their effects on the quality of life for affected individuals it is imperative that effective therapies are developed. Here we discuss some of the new approaches to CNS myelin repair that hold promise for reducing the burden of diseases characterized by myelin loss.
Collapse
Affiliation(s)
- Mohammad Abu-Rub
- Department of Neurology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| | - Robert H Miller
- Department of Anatomy and Regenerative Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|