Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38826423

ABSTRACT

Both neurons and glia communicate via diffusible neuromodulatory substances, but the substrates of computation in such neuromodulatory networks are unclear. During behavioral transitions in the larval zebrafish, the neuromodulator norepinephrine drives fast excitation and delayed inhibition of behavior and circuit activity. We find that the inhibitory arm of this feedforward motif is implemented by astroglial purinergic signaling. Neuromodulator imaging, behavioral pharmacology, and perturbations of neurons and astroglia reveal that norepinephrine triggers astroglial release of adenosine triphosphate, extracellular conversion into adenosine, and behavioral suppression through activation of hindbrain neuronal adenosine receptors. This work, along with a companion piece by Lefton and colleagues demonstrating an analogous pathway mediating the effect of norepinephrine on synaptic connectivity in mice, identifies a computational and behavioral role for an evolutionarily conserved astroglial purinergic signaling axis in norepinephrine-mediated behavioral and brain state transitions.

2.
Elife ; 122023 08 07.
Article in English | MEDLINE | ID: mdl-37548652

ABSTRACT

Sleep is a nearly universal feature of animal behaviour, yet many of the molecular, genetic, and neuronal substrates that orchestrate sleep/wake transitions lie undiscovered. Employing a viral insertion sleep screen in larval zebrafish, we identified a novel gene, dreammist (dmist), whose loss results in behavioural hyperactivity and reduced sleep at night. The neuronally expressed dmist gene is conserved across vertebrates and encodes a small single-pass transmembrane protein that is structurally similar to the Na+,K+-ATPase regulator, FXYD1/Phospholemman. Disruption of either fxyd1 or atp1a3a, a Na+,K+-ATPase alpha-3 subunit associated with several heritable movement disorders in humans, led to decreased night-time sleep. Since atpa1a3a and dmist mutants have elevated intracellular Na+ levels and non-additive effects on sleep amount at night, we propose that Dmist-dependent enhancement of Na+ pump function modulates neuronal excitability to maintain normal sleep behaviour.


Subject(s)
Sodium , Zebrafish , Animals , Humans , Zebrafish/genetics , Zebrafish/metabolism , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Homeostasis , Sleep/genetics , Phosphoproteins/metabolism
3.
PLoS One ; 18(2): e0278316, 2023.
Article in English | MEDLINE | ID: mdl-36757918

ABSTRACT

With the oncoming age of big data, biologists are encountering more use cases for cloud-based computing to streamline data processing and storage. Unfortunately, cloud platforms are difficult to learn, and there are few resources for biologists to demystify them. We have developed a guide for experimental biologists to set up cloud processing on Amazon Web Services to cheaply outsource data processing and storage. Here we provide a guide for setting up a computing environment in the cloud and showcase examples of using Python and Julia programming languages. We present example calcium imaging data in the zebrafish brain and corresponding analysis using suite2p software. Tools for budget and user management are further discussed in the attached protocol. Using this guide, researchers with limited coding experience can get started with cloud-based computing or move existing coding infrastructure into the cloud environment.


Subject(s)
Software , Zebrafish , Animals , Programming Languages , Information Storage and Retrieval , Cloud Computing
5.
Front Mol Neurosci ; 15: 873520, 2022.
Article in English | MEDLINE | ID: mdl-35465097

ABSTRACT

Sleep disorders and chronic sleep disturbances are common and are associated with cardio-metabolic diseases and neuropsychiatric disorders. Several genetic pathways and neuronal mechanisms that regulate sleep have been described in animal models, but the genes underlying human sleep variation and sleep disorders are largely unknown. Identifying these genes is essential in order to develop effective therapies for sleep disorders and their associated comorbidities. To address this unmet health problem, genome-wide association studies (GWAS) have identified numerous genetic variants associated with human sleep traits and sleep disorders. However, in most cases, it is unclear which gene is responsible for a sleep phenotype that is associated with a genetic variant. As a result, it is necessary to experimentally validate candidate genes identified by GWAS using an animal model. Rodents are ill-suited for this endeavor due to their poor amenability to high-throughput sleep assays and the high costs associated with generating, maintaining, and testing large numbers of mutant lines. Zebrafish (Danio rerio), an alternative vertebrate model for studying sleep, allows for the rapid and cost-effective generation of mutant lines using the CRISPR/Cas9 system. Numerous zebrafish mutant lines can then be tested in parallel using high-throughput behavioral assays to identify genes whose loss affects sleep. This process identifies a gene associated with each GWAS hit that is likely responsible for the human sleep phenotype. This strategy is a powerful complement to GWAS approaches and holds great promise to identify the genetic basis for common human sleep disorders.

6.
Mol Psychiatry ; 27(9): 3777-3793, 2022 09.
Article in English | MEDLINE | ID: mdl-35484242

ABSTRACT

Salient sensory stimuli are perceived by the brain, which guides both the timing and outcome of behaviors in a context-dependent manner. Light is such a stimulus, which is used in treating mood disorders often associated with a dysregulated hypothalamic-pituitary-adrenal stress axis. Relationships between the emotional valence of light and the hypothalamus, and how they interact to exert brain-wide impacts remain unclear. Employing larval zebrafish with analogous hypothalamic systems to mammals, we show in free-swimming animals that hypothalamic corticotropin releasing factor (CRFHy) neurons promote dark avoidance, and such role is not shared by other hypothalamic peptidergic neurons. Single-neuron projection analyses uncover processes extended by individual CRFHy neurons to multiple targets including sensorimotor and decision-making areas. In vivo calcium imaging uncovers a complex and heterogeneous response of individual CRFHy neurons to the light or dark stimulus, with a reduced overall sum of CRF neuronal activity in the presence of light. Brain-wide calcium imaging under alternating light/dark stimuli further identifies distinct and distributed photic response neuronal types. CRFHy neuronal ablation increases an overall representation of light in the brain and broadly enhances the functional connectivity associated with an exploratory brain state. These findings delineate brain-wide photic perception, uncover a previously unknown role of CRFHy neurons in regulating the perception and emotional valence of light, and suggest that light therapy may alleviate mood disorders through reducing an overall sum of CRF neuronal activity.


Subject(s)
Corticotropin-Releasing Hormone , Paraventricular Hypothalamic Nucleus , Animals , Corticotropin-Releasing Hormone/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Calcium , Zebrafish/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Brain/metabolism , Perception , Mammals/metabolism
7.
Bio Protoc ; 12(3): e4313, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35284597

ABSTRACT

Over the past decade, zebrafish have emerged as a powerful model for the study of vertebrate sleep and wake behaviors. Experimental evidence has demonstrated behavioral, anatomical, genetic, and pharmacological conservation of sleep between zebrafish and mammals, suggesting that discoveries in zebrafish can inform our understanding of mammalian sleep. Here, we describe a protocol for performing sleep behavioral experiments in larval zebrafish, using a high-throughput video tracking system. We explain how to set up a sleep behavioral experiment and provide guidelines on how to analyze the data. Using this protocol, a typical experiment can be completed in less than five days, and this method provides a scalable platform to perform genetic and pharmacological screens in a simple and cost-effective vertebrate model. By combining high-throughput behavioral assays with several advantageous features of zebrafish, this model system provides new opportunities to make discoveries that clarify the genetic and neurological mechanisms that regulate sleep.

8.
Hepatology ; 73(4): 1494-1508, 2021 04.
Article in English | MEDLINE | ID: mdl-32602149

ABSTRACT

BACKGROUND AND AIMS: The liver is a highly regenerative organ, but its regenerative capacity is compromised in severe liver injury settings. In chronic liver diseases, the number of liver progenitor cells (LPCs) correlates proportionally to disease severity, implying that their inefficient differentiation into hepatocytes exacerbates the disease. Moreover, LPCs secrete proinflammatory cytokines; thus, their prolonged presence worsens inflammation and induces fibrosis. Promoting LPC-to-hepatocyte differentiation in patients with advanced liver disease, for whom liver transplantation is currently the only therapeutic option, may be a feasible clinical approach because such promotion generates more functional hepatocytes and concomitantly reduces inflammation and fibrosis. APPROACH AND RESULTS: Here, using zebrafish models of LPC-mediated liver regeneration, we present a proof of principle of such therapeutics by demonstrating a role for the epidermal growth factor receptor (EGFR) signaling pathway in differentiation of LPCs into hepatocytes. We found that suppression of EGFR signaling promoted LPC-to-hepatocyte differentiation through the mitogen-activated ERK kinase (MEK)-extracellular signal-regulated kinase (ERK)-sex-determining region Y-box 9 (SOX9) cascade. Pharmacological inhibition of EGFR or MEK/ERK promoted LPC-to-hepatocyte differentiation as well as genetic suppression of the EGFR-ERK-SOX9 axis. Moreover, Sox9b overexpression in LPCs blocked their differentiation into hepatocytes. In the zebrafish liver injury model, both hepatocytes and biliary epithelial cells contributed to LPCs. EGFR inhibition promoted the differentiation of LPCs regardless of their origin. Notably, short-term treatment with EGFR inhibitors resulted in better liver recovery over the long term. CONCLUSIONS: The EGFR-ERK-SOX9 axis suppresses LPC-to-hepatocyte differentiation during LPC-mediated liver regeneration. We suggest EGFR inhibitors as a proregenerative therapeutic drug for patients with advanced liver disease.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Regeneration/drug effects , MAP Kinase Signaling System/drug effects , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Butadienes/pharmacology , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hepatocytes/cytology , Nitriles/pharmacology , Quinazolines/pharmacology , Stem Cells/cytology , Tyrphostins/pharmacology
9.
Elife ; 92020 12 18.
Article in English | MEDLINE | ID: mdl-33337320

ABSTRACT

Although several sleep-regulating neuronal populations have been identified, little is known about how they interact with each other to control sleep/wake states. We previously identified neuropeptide VF (NPVF) and the hypothalamic neurons that produce it as a sleep-promoting system (Lee et al., 2017). Here we show using zebrafish that npvf-expressing neurons control sleep via the serotonergic raphe nuclei (RN), a hindbrain structure that is critical for sleep in both diurnal zebrafish and nocturnal mice. Using genetic labeling and calcium imaging, we show that npvf-expressing neurons innervate and can activate serotonergic RN neurons. We also demonstrate that chemogenetic or optogenetic stimulation of npvf-expressing neurons induces sleep in a manner that requires NPVF and serotonin in the RN. Finally, we provide genetic evidence that NPVF acts upstream of serotonin in the RN to maintain normal sleep levels. These findings reveal a novel hypothalamic-hindbrain neuronal circuit for sleep/wake control.


Subject(s)
Hypothalamus/physiology , Neurons/metabolism , Neuropeptides/metabolism , Raphe Nuclei/metabolism , Sleep/physiology , Animals , Neural Pathways/physiology , Zebrafish
10.
Proc Natl Acad Sci U S A ; 117(44): 27400-27411, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33087579

ABSTRACT

Individual cell migration requires front-to-back polarity manifested by lamellipodial extension. At present, it remains debated whether and how membrane motility mediates this cell morphological change. To gain insights into these processes, we perform live imaging and molecular perturbation of migrating chick neural crest cells in vivo. Our results reveal an endocytic loop formed by circular membrane flow and anterograde movement of lipid vesicles, resulting in cell polarization and locomotion. Rather than clathrin-mediated endocytosis, macropinosomes encapsulate F-actin in the cell body, forming vesicles that translocate via microtubules to deliver actin to the anterior. In addition to previously proposed local conversion of actin monomers to polymers, we demonstrate a surprising role for shuttling of F-actin across cells for lamellipodial expansion. Thus, the membrane and cytoskeleton act in concert in distinct subcellular compartments to drive forward cell migration.


Subject(s)
Actins/metabolism , Cell Movement , Neural Crest/physiology , Pinocytosis , Pseudopodia/metabolism , Animals , Cell Membrane/metabolism , Chick Embryo , Intravital Microscopy , Neural Crest/cytology , Time-Lapse Imaging
11.
Sci Rep ; 10(1): 8352, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32415202

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

12.
Sci Rep ; 10(1): 7632, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32376893

ABSTRACT

Prokineticin receptors (PROKR1 and PROKR2) are G protein-coupled receptors which control human central and peripheral reproductive processes. Importantly, allelic variants of PROKR2 in humans are associated with altered migration of GnRH neurons, resulting in congenital hypogonadotropic hypogonadism (CHH), a heterogeneous disease characterized by delayed/absent puberty and/or infertility. Although this association is established in humans, murine models failed to fully recapitulate the reproductive and olfactory phenotypes observed in patients harboring PROKR2 mutations. Here, taking advantage of zebrafish model we investigated the role of prokr1b (ortholog of human PROKR2) during early stages of GnRH neuronal migration. Real-Time PCR and whole mount in situ hybridization assays indicate that prokr1b spatial-temporal expression is consistent with gnrh3. Moreover, knockdown and knockout of prokr1b altered the correct development of GnRH3 fibers, a phenotype that is rescued by injection of prokr1b mRNA. These results suggest that prokr1b regulates the development of the GnRH3 system in zebrafish. Analysis of gonads development and mating experiments indicate that prokr1b is not required for fertility in zebrafish, although its loss determine changes also at the testis level. Altogether, our results support the thesis of a divergent evolution in the control of vertebrate reproduction and provide a useful in vivo model for deciphering the mechanisms underlying the effect of PROKR2 allelic variants on CHH.

13.
Curr Biol ; 30(9): 1639-1648.e3, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32169212

ABSTRACT

Sleep is a cross-species phenomenon whose evolutionary and biological function remain poorly understood. Clinical and animal studies suggest that sleep disturbance is significantly associated with disruptions in protein homeostasis-or proteostasis-in the brain, but the mechanism of this link has not been explored. In the cell, the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) pathway modulates proteostasis by transiently inhibiting protein synthesis in response to proteostatic stress. In this study, we examined the role of the PERK pathway in sleep regulation and provide the first evidence that PERK signaling is required to regulate normal sleep in both vertebrates and invertebrates. We show that pharmacological inhibition of PERK reduces sleep in both Drosophila and zebrafish, indicating an evolutionarily conserved requirement for PERK in sleep. Genetic knockdown of PERK activity also reduces sleep in Drosophila, whereas PERK overexpression induces sleep. Finally, we demonstrate that changes in PERK signaling directly impact wake-promoting neuropeptide expression, revealing a mechanism through which proteostatic pathways can affect sleep and wake behavior. Taken together, these results demonstrate that protein synthesis pathways like PERK could represent a general mechanism of sleep and wake regulation and provide greater insight into the relationship between sleep and proteostasis.


Subject(s)
Biological Evolution , Sleep/genetics , Sleep/physiology , Zebrafish Proteins/metabolism , eIF-2 Kinase/metabolism , Animals , Cinnamates/pharmacology , Drosophila melanogaster , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Neuropeptides/genetics , Neuropeptides/metabolism , Signal Transduction , Thiourea/analogs & derivatives , Thiourea/pharmacology , Wakefulness/genetics , Wakefulness/physiology , Zebrafish , Zebrafish Proteins/genetics , eIF-2 Kinase/genetics
14.
Sci Adv ; 5(11): eaax4249, 2019 11.
Article in English | MEDLINE | ID: mdl-31763451

ABSTRACT

The genetic bases for most human sleep disorders and for variation in human sleep quantity and quality are largely unknown. Using the zebrafish, a diurnal vertebrate, to investigate the genetic regulation of sleep, we found that epidermal growth factor receptor (EGFR) signaling is necessary and sufficient for normal sleep levels and is required for the normal homeostatic response to sleep deprivation. We observed that EGFR signaling promotes sleep via mitogen-activated protein kinase/extracellular signal-regulated kinase and RFamide neuropeptide signaling and that it regulates RFamide neuropeptide expression and neuronal activity. Consistent with these findings, analysis of a large cohort of human genetic data from participants of European ancestry revealed that common variants in genes within the EGFR signaling pathway are associated with variation in human sleep quantity and quality. These results indicate that EGFR signaling and its downstream pathways play a central and ancient role in regulating sleep and provide new therapeutic targets for sleep disorders.


Subject(s)
ErbB Receptors/genetics , Gene Expression Regulation , Neuropeptides/genetics , Signal Transduction/genetics , Sleep/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Evolution, Molecular , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Mitogen-Activated Protein Kinases/genetics
15.
Sci Rep ; 9(1): 12405, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455847

ABSTRACT

The circadian clock ensures that behavioral and physiological processes occur at appropriate times during the 24-hour day/night cycle, and is regulated at both the cellular and organismal levels. To identify pathways acting on intact animals, we performed a small molecule screen using a luminescent reporter of molecular circadian rhythms in zebrafish larvae. We identified both known and novel pathways that affect circadian period, amplitude and phase. Several drugs identified in the screen did not affect circadian rhythms in cultured cells derived from luminescent reporter embryos or in established zebrafish and mammalian cell lines, suggesting they act via mechanisms absent in cell culture. Strikingly, using drugs that promote or inhibit inflammation, as well as a mutant that lacks microglia, we found that inflammatory state affects circadian amplitude. These results demonstrate a benefit of performing drug screens using intact animals and provide novel targets for treating circadian rhythm disorders.


Subject(s)
Circadian Rhythm/drug effects , Small Molecule Libraries/pharmacology , Zebrafish/physiology , Animals , Animals, Genetically Modified/physiology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Casein Kinase I/antagonists & inhibitors , Casein Kinase I/metabolism , Larva/drug effects , Larva/physiology , Protein Kinase Inhibitors/pharmacology , Receptors, Glycine/agonists , Receptors, Glycine/metabolism , Taurine/pharmacology , Zebrafish/growth & development , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/metabolism
16.
Cell ; 178(4): 850-866.e26, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31398340

ABSTRACT

We performed a comprehensive assessment of rare inherited variation in autism spectrum disorder (ASD) by analyzing whole-genome sequences of 2,308 individuals from families with multiple affected children. We implicate 69 genes in ASD risk, including 24 passing genome-wide Bonferroni correction and 16 new ASD risk genes, most supported by rare inherited variants, a substantial extension of previous findings. Biological pathways enriched for genes harboring inherited variants represent cytoskeletal organization and ion transport, which are distinct from pathways implicated in previous studies. Nevertheless, the de novo and inherited genes contribute to a common protein-protein interaction network. We also identified structural variants (SVs) affecting non-coding regions, implicating recurrent deletions in the promoters of DLG2 and NR3C2. Loss of nr3c2 function in zebrafish disrupts sleep and social function, overlapping with human ASD-related phenotypes. These data support the utility of studying multiplex families in ASD and are available through the Hartwell Autism Research and Technology portal.


Subject(s)
Autism Spectrum Disorder/genetics , Genetic Predisposition to Disease/genetics , Pedigree , Protein Interaction Maps/genetics , Animals , Child , Databases, Genetic , Disease Models, Animal , Female , Gene Deletion , Guanylate Kinases/genetics , Humans , Inheritance Patterns/genetics , Machine Learning , Male , Nuclear Family , Promoter Regions, Genetic/genetics , Receptors, Mineralocorticoid/genetics , Risk Factors , Tumor Suppressor Proteins/genetics , Whole Genome Sequencing , Zebrafish/genetics
17.
Cell Rep ; 27(10): 2871-2880.e2, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31167134

ABSTRACT

Retinal waves, the spontaneous patterned neural activities propagating among developing retinal ganglion cells (RGCs), instruct the activity-dependent refinement of visuotopic maps. Although it is known that the wave is initiated successively by amacrine cells and bipolar cells, the behavior and function of glia in retinal waves remain unclear. Using multiple in vivo methods in larval zebrafish, we found that Müller glial cells (MGCs) display wave-like spontaneous activities, which start at MGC processes within the inner plexiform layer, vertically spread to their somata and endfeet, and horizontally propagate into neighboring MGCs. MGC waves depend on glutamatergic signaling derived from bipolar cells. Moreover, MGCs express both glia-specific glutamate transporters and the AMPA subtype of glutamate receptors. The AMPA receptors mediate MGC calcium activities during retinal waves, whereas the glutamate transporters modulate the occurrence of retinal waves. Thus, MGCs can sense and regulate retinal waves via AMPA receptors and glutamate transporters, respectively.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Calcium/metabolism , Ependymoglial Cells/metabolism , Glutamic Acid/metabolism , Neuroglia/metabolism , Receptors, AMPA/metabolism , Retinal Ganglion Cells/metabolism , Amacrine Cells/metabolism , Amacrine Cells/physiology , Amino Acid Transport System X-AG/antagonists & inhibitors , Amino Acid Transport System X-AG/genetics , Animals , Animals, Genetically Modified , Ependymoglial Cells/cytology , Ependymoglial Cells/drug effects , Ependymoglial Cells/physiology , Glutamic Acid/pharmacology , Larva/drug effects , Larva/metabolism , Larva/physiology , Neuroglia/cytology , Neuroglia/physiology , Receptors, AMPA/antagonists & inhibitors , Retina/cytology , Retina/metabolism , Retina/physiology , Retinal Bipolar Cells/metabolism , Retinal Bipolar Cells/physiology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/physiology , Zebrafish
18.
Neuron ; 103(4): 686-701.e8, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31248729

ABSTRACT

The role of serotonin (5-HT) in sleep is controversial: early studies suggested a sleep-promoting role, but eventually the paradigm shifted toward a wake-promoting function for the serotonergic raphe. Here, we provide evidence from zebrafish and mice that the raphe are critical for the initiation and maintenance of sleep. In zebrafish, genetic ablation of 5-HT production by the raphe reduces sleep, sleep depth, and the homeostatic response to sleep deprivation. Pharmacological inhibition or ablation of the raphe reduces sleep, while optogenetic stimulation increases sleep. Similarly, in mice, ablation of the raphe increases wakefulness and impairs the homeostatic response to sleep deprivation, whereas tonic optogenetic stimulation at a rate similar to baseline activity induces sleep. Interestingly, burst optogenetic stimulation induces wakefulness in accordance with previously described burst activity of the raphe during arousing stimuli. These results indicate that the serotonergic system promotes sleep in both diurnal zebrafish and nocturnal rodents. VIDEO ABSTRACT.


Subject(s)
Mice/physiology , Raphe Nuclei/physiology , Serotonin/physiology , Sleep/physiology , Zebrafish/physiology , Animals , Arousal/genetics , Arousal/physiology , Buspirone/pharmacology , Circadian Rhythm/physiology , Fenclonine/pharmacology , Homeostasis , Male , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics , Quipazine/pharmacology , Serotonergic Neurons/drug effects , Serotonergic Neurons/physiology , Serotonin/biosynthesis , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Sleep Deprivation/genetics , Sleep Deprivation/physiopathology , Tryptophan Hydroxylase/deficiency , Tryptophan Hydroxylase/genetics , Wakefulness/genetics , Wakefulness/physiology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
19.
Cell Rep ; 27(1): 115-128.e5, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30943395

ABSTRACT

During development, oligodendrocyte progenitor cells (OPCs) migrate extensively throughout the spinal cord. However, their migration is restricted at transition zones (TZs). At these specialized locations, unique glial cells in both zebrafish and mice play a role in preventing peripheral OPC migration, but the mechanisms of this regulation are not understood. To elucidate the mechanisms that mediate OPC segregation at motor exit point (MEP) TZs, we performed an unbiased small-molecule screen. Using chemical screening and in vivo imaging, we discovered that inhibition of A2a adenosine receptors (ARs) causes ectopic OPC migration out of the spinal cord. We provide in vivo evidence that neuromodulation, partially mediated by adenosine, influences OPC migration specifically at the MEP TZ. This work opens exciting possibilities for understanding how OPCs reach their final destinations during development and identifies mechanisms that could promote their migration in disease.


Subject(s)
Adenosine/pharmacology , Cell Movement/drug effects , Motor Endplate/embryology , Neurotransmitter Agents/pharmacology , Oligodendroglia/drug effects , Spinal Cord/embryology , Animals , Animals, Genetically Modified , Body Patterning/physiology , Cell Differentiation/drug effects , Embryo, Nonmammalian , Female , Male , Motor Endplate/cytology , Motor Neurons/drug effects , Motor Neurons/physiology , Oligodendroglia/physiology , Spinal Cord/cytology , Spinal Cord/drug effects , Stem Cells/drug effects , Stem Cells/physiology , Zebrafish/embryology
20.
Science ; 363(6426): 455-456, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30705172
SELECTION OF CITATIONS
SEARCH DETAIL
...