Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.094
Filter
1.
J Neurosci ; 44(40)2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358028

ABSTRACT

The brain is a highly adaptable organ that is molded by experience throughout life. Although the field of neuroscience has historically focused on intrinsic neuronal mechanisms of plasticity, there is growing evidence that multiple glial populations regulate the timing and extent of neuronal plasticity, particularly over the course of development. This review highlights recent discoveries on the role of glial cells in the establishment of cortical circuits and the regulation of experience-dependent neuronal plasticity during critical periods of neurodevelopment. These studies provide strong evidence that neuronal circuit maturation and plasticity are non-cell autonomous processes that require both glial-neuronal and glial-glial cross talk to proceed. We conclude by discussing open questions that will continue to guide research in this nascent field.


Subject(s)
Cerebral Cortex , Neuroglia , Neuronal Plasticity , Neurons , Neuronal Plasticity/physiology , Animals , Neuroglia/physiology , Humans , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Neurons/physiology , Nerve Net/physiology , Nerve Net/growth & development , Neurogenesis/physiology
2.
J Neurosci ; 44(40)2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358029

ABSTRACT

Communication between neurons and glia significantly influences the development maturation, plasticity, and disease progressions of the nervous system. As a new signaling modality, extracellular vesicles display a diverse role for robust functional regulation of neurons through their protein and nucleic acid cargoes. This review highlights recent breakthroughs in the research of signaling mechanisms between glia and neurons mediated by extracellular vesicles that are important for neural development, axonal maintenance, synaptic functions, and disease progression in the mammalian nervous system. We will discuss the biological roles of extracellular vesicles released from neurons, astroglia, microglia, and oligodendroglia in the nervous system and their implications in neurodegenerative disorders.


Subject(s)
Cell Communication , Central Nervous System , Extracellular Vesicles , Neuroglia , Neurons , Extracellular Vesicles/physiology , Extracellular Vesicles/metabolism , Humans , Animals , Neurons/physiology , Neuroglia/physiology , Cell Communication/physiology , Central Nervous System/physiology , Central Nervous System/cytology
3.
J Neurosci ; 44(40)2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358030

ABSTRACT

The traditional view of glial cells as mere supportive tissue has shifted, due to advances in technology and theoretical conceptualization, to include a diversity of other functions, such as regulation of complex behaviors. Astrocytes, the most abundant glial cells in the central nervous system (CNS), have been shown to modulate synaptic functions through gliotransmitter-mediated neurotransmitter reuptake, influencing neuronal signaling and behavioral functions. Contemporary studies further highlight astrocytes' involvement in complex cognitive functions. For instance, inhibiting astrocytes in the hippocampus can lead to memory deficits, suggesting their integral role in memory processes. Moreover, astrocytic calcium activity and astrocyte-neuron metabolic coupling have been linked to changes in synaptic strength and learning. Microglia, another type of glial cell, also extend beyond their supportive roles, contributing to learning and memory processes, with microglial reductions impacting these functions in a developmentally dependent manner. Oligodendrocytes, traditionally thought to have limited roles postdevelopment, are now recognized for their activity-dependent modulation of myelination and plasticity, thus influencing behavioral responses. Recent advancements in technology and computational modeling have expanded our understanding of glial functions, particularly how astrocytes influence neuronal circuits and behaviors. This review underscores the importance of glial cells in CNS functions and the need for further research to unravel the complexities of neuron-glia interactions, the impact of these interactions on brain functions, and potential implications for neurological diseases.


Subject(s)
Cognition , Neuroglia , Neuronal Plasticity , Neurons , Neuronal Plasticity/physiology , Animals , Humans , Neuroglia/physiology , Neurons/physiology , Cognition/physiology , Cell Communication/physiology , Astrocytes/physiology
4.
Commun Biol ; 7(1): 1286, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39384971

ABSTRACT

Bidirectional communication between neurons and glial cells is crucial to establishing and maintaining normal brain function. Some of these interactions are activity-dependent, yet it remains largely unexplored how acute changes in neuronal activity affect glial-to-neuron and neuron-to-glial dynamics. Here, we use excitatory and inhibitory designer receptors exclusively activated by designer drugs (DREADD) to study the effects of acute chemogenetic manipulations of a subpopulation of layer 5 cortical projection and dentate gyrus neurons in adult (Rbp4Cre) mouse brains. We show that acute chemogenetic neuronal activation reduces synaptic density, and increases microglia and astrocyte reactivity, but does not affect parvalbumin (PV+) neurons, only perineuronal nets (PNN). Conversely, acute silencing increases synaptic density and decreases glial reactivity. We show fast glial response upon clozapine-N-oxide (CNO) administration in cortical and subcortical regions. Together, our work provides evidence of fast, activity-dependent, bidirectional interactions between neurons and glial cells.


Subject(s)
Clozapine , Neuroglia , Neurons , Animals , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Neuroglia/metabolism , Neuroglia/drug effects , Neuroglia/physiology , Mice , Clozapine/pharmacology , Clozapine/analogs & derivatives , Male , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Parvalbumins/metabolism , Mice, Transgenic
5.
Sci Rep ; 14(1): 22282, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333287

ABSTRACT

Infrared neural stimulation (INS) emerges as a promising tool for stimulating the nervous system by its high spatial precision and absence of the use of exogenous agents into the tissue, which led to the first successful proof of concept in human brain. While neural networks have been the focal point of INS research, this technique is also non cell type specific as it triggers activity in non electrically excitable cells. Despite increasing interest, there remains to demonstrate well defined simultaneous astrocytic and neuronal signals in response to INS. Using calcium imaging, we show that INS has the capacity to initiate calcium signaling in both astrocytes and neurons simultaneously from the rostral lumbar spinal cord, each exhibiting distinct temporal and amplitude characteristics. Importantly, the mechanism underlying infrared-induced neuronal and astrocytic calcium signaling differ, with neuronal activity relying on sodium channels, whereas induced astrocytic signaling is predominantly influenced by extracellular calcium and TRPV4 channels. Furthermore, our findings demonstrate the frequency shift of neuronal calcium oscillations through infrared stimulation. By deepening our understanding in INS fundamentals, this technique holds great promise for advancing neuroscience, deepening our understanding of pathologies, and potentially paving the way for future clinical applications.


Subject(s)
Astrocytes , Calcium Signaling , Infrared Rays , Neurons , Spinal Cord , Astrocytes/metabolism , Spinal Cord/physiology , Spinal Cord/metabolism , Animals , Neurons/metabolism , Neurons/physiology , Calcium/metabolism , TRPV Cation Channels/metabolism , Locomotion/physiology , Nerve Net/physiology , Mice , Neuroglia/metabolism , Neuroglia/physiology
6.
Neuron ; 112(18): 2993-2995, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39326387

ABSTRACT

While the enteric nervous system (ENS) is highly dynamic during development, the extent to which it is capable of repair remains unclear. In this issue of Neuron, Stavely et al.1 show that enteric neurons can reinnervate damaged regions to regain functionality using a glial positioning system (GPS) as their guide.


Subject(s)
Enteric Nervous System , Nerve Regeneration , Neuroglia , Neuroglia/physiology , Enteric Nervous System/physiology , Enteric Nervous System/cytology , Animals , Nerve Regeneration/physiology , Neurites/physiology , Intestines/physiology , Humans
7.
Front Immunol ; 15: 1393842, 2024.
Article in English | MEDLINE | ID: mdl-39136008

ABSTRACT

Chondroitin sulfate proteoglycans (CSPGs) are fundamental components of the extracellular matrix in the central nervous system (CNS). Among these, the Nerve-Glial antigen 2 (NG2) stands out as a transmembrane CSPG exclusively expressed in a different population of cells collectively termed NG2-expressing cells. These enigmatic cells, found throughout the developing and adult CNS, have been indicated with various names, including NG2 progenitor cells, polydendrocytes, synantocytes, NG2 cells, and NG2-Glia, but are more commonly referred to as oligodendrocyte progenitor cells. Characterized by high proliferation rates and unique morphology, NG2-expressing cells stand apart from neurons, astrocytes, and oligodendrocytes. Intriguingly, some NG2-expressing cells form functional glutamatergic synapses with neurons, challenging the long-held belief that only neurons possess the intricate machinery required for neurotransmission. In the CNS, the complexity surrounding NG2-expressing cells extends to their classification. Additionally, NG2 expression has been documented in pericytes and immune cells, suggesting a role in regulating brain innate immunity and neuro-immune crosstalk in homeostasis. Ongoing debates revolve around their heterogeneity, potential as progenitors for various cell types, responses to neuroinflammation, and the role of NG2. Therefore, this review aims to shed light on the enigma of NG2-expressing cells by delving into their structure, functions, and signaling pathways. We will critically evaluate the literature on NG2 expression across the CNS, and address the contentious issues surrounding their classification and roles in neuroinflammation and neurodegeneration. By unraveling the intricacies of NG2-expressing cells, we hope to pave the way for a more comprehensive understanding of their contributions to CNS health and during neurological disorders.


Subject(s)
Antigens , Central Nervous System , Humans , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Antigens/immunology , Antigens/metabolism , Neuroglia/metabolism , Neuroglia/immunology , Neuroglia/physiology , Neurons/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Proteoglycans
8.
Eur J Neurosci ; 60(5): 5019-5039, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39099396

ABSTRACT

Modulation of input from primary afferent fibres has long been examined at the level of the first relays of these fibres. However, recent studies reveal that input to the spinal cord may also be modulated at the level of the very entry of afferent fibres to the spinal grey matter before action potentials in intraspinal collaterals of afferent fibres reach their target neurons. Such modulation greatly depends on the actions of GABA via extrasynaptic membrane receptors. In the reported study we hypothesized that the increase in excitability of afferent fibres following epidural polarization close to the site where collaterals of afferent fibres leave the dorsal columns is due to the release of GABA from two sources: not only GABAergic interneurons but also glial cells. We present evidence, primo, that GABA released from both these sources contributes to a long-lasting increase in the excitability and a shortening of the refractory period of epidurally stimulated afferent fibres and, secondo, that effects of epidural polarization on the release of GABA are more critical for these changes than direct effects of DC on the stimulated fibres. The experiments were carried out in deeply anaesthetized rats in which changes in compound action potentials evoked in hindlimb peripheral nerves by dorsal column stimulation were used as a measure of the excitability of afferent fibres. The study throws new light on the modulation of input to spinal networks but also on mechanisms underlying the restoration of spinal functions.


Subject(s)
Interneurons , Neuroglia , Spinal Cord , gamma-Aminobutyric Acid , Animals , Interneurons/metabolism , Interneurons/physiology , Spinal Cord/metabolism , Spinal Cord/physiology , Rats , gamma-Aminobutyric Acid/metabolism , Neuroglia/metabolism , Neuroglia/physiology , Male , Action Potentials/physiology , Epidural Space/physiology , Electric Stimulation , Rats, Wistar , Rats, Sprague-Dawley , Afferent Pathways/physiology , Afferent Pathways/metabolism
9.
Eur J Neurosci ; 60(6): 5156-5168, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39126378

ABSTRACT

The subventricular zone (SVZ) is one of the neurogenic regions of the adult mammalian brain. Neural stem cells (NSCs) in the SVZ have certain key features: they express glial fibrillary acidic protein (GFAP), proliferate slowly, have a radial glia-like (RG-L) morphology, and are in contact with the cerebrospinal fluid (CSF). NSCs have been isolated by FACS to analyse them, but their morphology has not been systematically examined. To address this knowledge gap, we sparsely labelled RG-L cells in the SVZ of neonatal mice by introducing via electroporation a plasmid expressing fluorescent protein under the control of the GFAP promoter. We then classified RG-L cells into three types (RG-L1, 2, and 3) based on their morphologies. RG-L1 cells had a basal process with some branches and numerous fine processes. RG-L2 cells had a basal process, but fewer branches and fine processes than RG-L1 cells. RG-L3 cells had one basal process that was almost free of branches and fine processes. Importantly, regardless of the cell type, about half of their somata resided on the basal side of the SVZ. Based on changes in their proportions during postnatal development and their expression of GFAP and cell proliferation markers at the adult stage, we speculated that NSCs change their morphologies during development/maturation and not all NSCs must always be in the apical SVZ or in contact with the CSF. Our results indicate that in addition to expression of markers for NSCs, the morphology is a critical feature to identify NSCs.


Subject(s)
Glial Fibrillary Acidic Protein , Lateral Ventricles , Neural Stem Cells , Animals , Mice , Lateral Ventricles/cytology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Animals, Newborn , Neuroglia/cytology , Neuroglia/physiology , Neuroglia/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/physiology , Ependymoglial Cells/metabolism , Cell Proliferation/physiology
10.
Nat Neurosci ; 27(8): 1427, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107594
11.
Poult Sci ; 103(10): 104070, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094494

ABSTRACT

Enteric glial cell (EGC) is involved in neuroimmune regulation within the enteric nervous system (ENS); however, limited information exists on the distribution and ultrastructure of EGC in the poultry gut. We aim to investigate the morphological features and distribution of EGC in the chicken cecum. Here, we investigated the distribution and ultrastructural features of chicken cecum EGC using immunohistochemistry (IHC) and transmission electron microscopy (TEM). IHC showed that EGC was widely distributed throughout the chicken cecum. In the mucosal layer, EGC was morphologically irregular, with occasionally interconnecting protrusions that outlined signal-negative neurons. The morphology of EGC in the submucosal layer was also irregular. In the inner circular muscle layer and between the inner circular and outer longitudinal muscle layers, EGC aligned parallel to the circular muscle cells. A small number of EGC with an irregular morphology were found in the outer longitudinal muscle layer. In addition, in the submucosal and myenteric plexus, EGC were aggregated, and the protrusions of the immunoreactive cells interconnected to outline the bodies of nonreactive neurons. TEM-guided ultrastructural characterization confirmed the IHC findings that EGC were morphologically irregular and revealed they developed either a star, bipolar, or fibrous shape. The nucleus was also irregular, with electron-dense heterochromatin distributed in the center of the nucleus or on the nuclear membrane. The cytoplasm contained many glial filaments and vesicle-containing protrusions from neuronal cells; organelles were rare. EGC was in close contact with other cells in their vicinity. These findings suggest that EGC is well-situated to exert influence on intestinal motility and immune functions through mechanical contraction and chemical secretion.


Subject(s)
Cecum , Chickens , Enteric Nervous System , Microscopy, Electron, Transmission , Neuroglia , Animals , Neuroglia/ultrastructure , Neuroglia/physiology , Cecum/ultrastructure , Microscopy, Electron, Transmission/veterinary , Enteric Nervous System/ultrastructure , Enteric Nervous System/physiology , Immunohistochemistry/veterinary , Male
12.
J Neurophysiol ; 132(3): 1056-1073, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39110896

ABSTRACT

Besides having high potency and efficacy at the µ-opioid (MOR) and other opioid receptor types, fentanyl has some affinity for some adrenergic receptor types, which may underlie its unique pathophysiological differences from typical opioids. To better understand the unique actions of fentanyl, we assessed the extent to which fentanyl alters striatal medium spiny neuron (MSN) activity via opioid receptors or α1-adrenoceptors in dopamine type 1 or type 2 receptor (D1 or D2)-expressing MSNs. In neuronal and mixed-glial cocultures from the striatum, acute fentanyl (100 nM) exposure decreased the frequency of spontaneous action potentials. Overnight exposure of cocultures to 100 nM fentanyl severely reduced the proportion of MSNs with spontaneous action potentials, which was unaffected by coexposure to the opioid receptor antagonist naloxone (10 µM) but fully negated by coadministering the pan-α1-adrenoceptor inverse agonist prazosin (100 nM) and partially reversed by the selective α1A-adrenoceptor antagonist RS 100329 (300 nM). Acute fentanyl (100 nM) exposure modestly reduced the frequency of action potentials and caused firing rate adaptations in D2, but not D1, MSNs. Prolonged (2-5 h) fentanyl (100 nM) application dramatically attenuated firing rates in both D1 and D2 MSNs. To identify possible cellular sites of α1-adrenoceptor action, α1-adrenoceptors were localized in subpopulations of striatal astroglia and neurons by immunocytochemistry and Adra1a mRNA by in situ hybridization in astrocytes. Thus, sustained fentanyl exposure can inhibit striatal MSN activity via a nonopioid receptor-dependent pathway, which may be modulated via complex actions in α1-adrenoceptor-expressing striatal neurons and/or glia.NEW & NOTEWORTHY Acute fentanyl exposure attenuated the activity of striatal medium spiny neurons (MSNs) in vitro and in dopamine D2, but not D1, receptor-expressing MSNs in ex vivo slices. By contrast, sustained fentanyl exposure suppressed the spontaneous activity of MSNs cocultured with glia through a nonopioid receptor-dependent mechanism modulated, in part, by α1-adrenoceptors. Fentanyl exposure can affect striatal function via a nonopioid receptor mechanism of action that appears mediated by α1-adrenoreceptor-expressing striatal neurons and/or astroglia.


Subject(s)
Action Potentials , Analgesics, Opioid , Coculture Techniques , Corpus Striatum , Fentanyl , Neuroglia , Neurons , Animals , Fentanyl/pharmacology , Neuroglia/drug effects , Neuroglia/physiology , Neuroglia/metabolism , Neurons/drug effects , Neurons/physiology , Mice , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiology , Analgesics, Opioid/pharmacology , Action Potentials/drug effects , Receptors, Opioid/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D2/drug effects , Male , Narcotic Antagonists/pharmacology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Cells, Cultured
13.
Glia ; 72(10): 1766-1784, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39141572

ABSTRACT

The central and peripheral nervous systems (CNS and PNS, respectively) exhibit remarkable diversity in the capacity to regenerate following neuronal injury with PNS injuries being much more likely to regenerate than those that occur in the CNS. Glial responses to damage greatly influence the likelihood of regeneration by either promoting or inhibiting axonal regrowth over time. However, despite our understanding of how some glial lineages participate in nerve degeneration and regeneration, less is known about the contributions of peripheral satellite glial cells (SGC) to regeneration failure following central axon branch injury of dorsal root ganglia (DRG) sensory neurons. Here, using in vivo, time-lapse imaging in larval zebrafish coupled with laser axotomy, we investigate the role of SGCs in axonal regeneration. In our studies we show that SGCs respond to injury by relocating their nuclei to the injury site during the same period that DRG neurons produce new central branch neurites. Laser ablation of SGCs prior to axon injury results in more neurite growth attempts and ultimately a higher rate of successful central axon regrowth, implicating SGCs as inhibitors of regeneration. We also demonstrate that this SGC response is mediated in part by ErbB signaling, as chemical inhibition of this receptor results in reduced SGC motility and enhanced central axon regrowth. These findings provide new insights into SGC-neuron interactions under injury conditions and how these interactions influence nervous system repair.


Subject(s)
Axotomy , Ganglia, Spinal , Nerve Regeneration , Zebrafish , Animals , Nerve Regeneration/physiology , Animals, Genetically Modified , Spinal Cord , Satellite Cells, Perineuronal/physiology , Neuroglia/physiology , Zebrafish Proteins/metabolism , Axons/physiology
14.
Front Immunol ; 15: 1408744, 2024.
Article in English | MEDLINE | ID: mdl-38957473

ABSTRACT

Enteric glial cells (EGCs) are an essential component of the enteric nervous system (ENS) and play key roles in gastrointestinal development, homeostasis, and disease. Derived from neural crest cells, EGCs undergo complex differentiation processes regulated by various signalling pathways. Being among the most dynamic cells of the digestive system, EGCs react to cues in their surrounding microenvironment and communicate with various cell types and systems within the gut. Morphological studies and recent single cell RNA sequencing studies have unveiled heterogeneity among EGC populations with implications for regional functions and roles in diseases. In gastrointestinal disorders, including inflammatory bowel disease (IBD), infections and cancer, EGCs modulate neuroplasticity, immune responses and tumorigenesis. Recent evidence suggests that EGCs respond plastically to the microenvironmental cues, adapting their phenotype and functions in disease states and taking on a crucial role. They exhibit molecular abnormalities and alter communication with other intestinal cell types, underscoring their therapeutic potential as targets. This review delves into the multifaceted roles of EGCs, particularly emphasizing their interactions with various cell types in the gut and their significant contributions to gastrointestinal disorders. Understanding the complex roles of EGCs in gastrointestinal physiology and pathology will be crucial for the development of novel therapeutic strategies for gastrointestinal disorders.


Subject(s)
Enteric Nervous System , Neuroglia , Humans , Neuroglia/physiology , Enteric Nervous System/pathology , Animals , Gastrointestinal Diseases/pathology
15.
Nat Rev Neurosci ; 25(8): 519-534, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38951687

ABSTRACT

During central nervous system (CNS) development, neural progenitor cells (NPCs) generate neurons and glia in two different ways. In direct neurogenesis, daughter cells differentiate directly into neurons or glia, whereas in indirect neurogenesis, neurons or glia are generated after one or more daughter cell divisions. Intriguingly, indirect neurogenesis is not stochastically deployed and plays instructive roles during CNS development: increased generation of cells from specific lineages; increased generation of early or late-born cell types within a lineage; and increased cell diversification. Increased indirect neurogenesis might contribute to the anterior CNS expansion evident throughout the Bilateria and help to modify brain-region size without requiring increased NPC numbers or extended neurogenesis. Increased indirect neurogenesis could be an evolutionary driver of the gyrencephalic (that is, folded) cortex that emerged during mammalian evolution and might even have increased during hominid evolution. Thus, selection of indirect versus direct neurogenesis provides a powerful developmental and evolutionary instrument that drives not only the evolution of CNS complexity but also brain expansion and modulation of brain-region size, and thereby the evolution of increasingly advanced cognitive abilities. This Review describes indirect neurogenesis in several model species and humans, and highlights some of the molecular genetic mechanisms that control this important process.


Subject(s)
Neurogenesis , Neurogenesis/physiology , Humans , Animals , Biological Evolution , Neural Stem Cells/physiology , Neural Stem Cells/cytology , Neurons/physiology , Cell Differentiation/physiology , Central Nervous System/physiology , Central Nervous System/growth & development , Central Nervous System/cytology , Neuroglia/physiology , Brain/physiology , Brain/growth & development , Brain/cytology
16.
Bioessays ; 46(9): e2400133, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38990084

ABSTRACT

The vertebrate retina is a tractable system for studying control of cell neurogenesis and cell fate specification. During embryonic development, retinal neurogenesis is under strict temporal regulation, with cell types generated in fixed but overlapping temporal intervals. The temporal sequence and relative numbers of retinal cell types generated during development are robust and show minimal experience-dependent variation. In many cold-blooded vertebrates, acute retinal injury induces a different form of neurogenesis, where Müller glia reprogram into retinal progenitor-like cells that selectively regenerate retinal neurons lost to injury. The extent to which the molecular mechanisms controlling developmental and injury-induced neurogenesis resemble one another has long been unclear. However, a recent study in zebrafish has shed new light on this question, using single-cell multiomic analysis to show that selective loss of different retinal cell types induces the formation of fate-restricted Müller glia-derived progenitors that differ both from one another and from progenitors in developing retina. Here, we discuss the broader implications of these findings, and their possible therapeutic relevance.


Subject(s)
Neurogenesis , Retina , Zebrafish , Animals , Ependymoglial Cells/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/physiology , Humans , Regeneration/physiology , Cell Differentiation , Neuroglia/metabolism , Neuroglia/physiology
17.
Neuron ; 112(18): 3143-3160.e6, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39019043

ABSTRACT

Here, we establish that plasticity exists within the postnatal enteric nervous system by demonstrating the reinnervation potential of post-mitotic enteric neurons (ENs). Employing BAF53b-Cre mice for selective neuronal tracing, the reinnervation capabilities of mature postnatal ENs are shown across multiple model systems. Isolated ENs regenerate neurites in vitro, with neurite complexity and direction influenced by contact with enteric glial cells (EGCs). Nerve fibers from transplanted ENs exclusively interface and travel along EGCs within the muscularis propria. Resident EGCs persist after Cre-dependent ablation of ENs and govern the architecture of the myenteric plexus for reinnervating ENs, as shown by nerve fiber projection tracing. Transplantation and optogenetic experiments in vivo highlight the rapid reinnervation potential of post-mitotic neurons, leading to restored gut muscle contractile activity within 2 weeks. These studies illustrate the structural and functional reinnervation capacity of post-mitotic ENs and the critical role of EGCs in guiding and patterning their trajectories.


Subject(s)
Enteric Nervous System , Neuroglia , Neurons , Animals , Neuroglia/physiology , Enteric Nervous System/physiology , Enteric Nervous System/cytology , Mice , Neurons/physiology , Intestines/innervation , Intestines/physiology , Nerve Regeneration/physiology , Myenteric Plexus/cytology , Myenteric Plexus/physiology , Mice, Transgenic , Neurites/physiology
18.
Learn Mem ; 31(5)2024 May.
Article in English | MEDLINE | ID: mdl-38862167

ABSTRACT

Providing metabolic support to neurons is now recognized as a major function of glial cells that is conserved from invertebrates to vertebrates. However, research in this field has focused for more than two decades on the relevance of lactate and glial glycolysis for neuronal energy metabolism, while overlooking many other facets of glial metabolism and their impact on neuronal physiology, circuit activity, and behavior. Here, we review recent work that has unveiled new features of glial metabolism, especially in Drosophila, in the modulation of behavioral traits involving the mushroom bodies (MBs). These recent findings reveal that spatially and biochemically distinct modes of glucose-derived neuronal fueling are implemented within the MB in a memory type-specific manner. In addition, cortex glia are endowed with several antioxidant functions, whereas astrocytes can serve as pro-oxidant agents that are beneficial to redox signaling underlying long-term memory. Finally, glial fatty acid oxidation seems to play a dual fail-safe role: first, as a mode of energy production upon glucose shortage, and, second, as a factor underlying the clearance of excessive oxidative load during sleep. Altogether, these integrated studies performed in Drosophila indicate that glial metabolism has a deterministic role on behavior.


Subject(s)
Behavior, Animal , Mushroom Bodies , Neuroglia , Animals , Mushroom Bodies/metabolism , Mushroom Bodies/physiology , Neuroglia/metabolism , Neuroglia/physiology , Behavior, Animal/physiology , Drosophila , Energy Metabolism/physiology
20.
Elife ; 132024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905123

ABSTRACT

The brain is consisted of diverse neurons arising from a limited number of neural stem cells. Drosophila neural stem cells called neuroblasts (NBs) produces specific neural lineages of various lineage sizes depending on their location in the brain. In the Drosophila visual processing centre - the optic lobes (OLs), medulla NBs derived from the neuroepithelium (NE) give rise to neurons and glia cells of the medulla cortex. The timing and the mechanisms responsible for the cessation of medulla NBs are so far not known. In this study, we show that the termination of medulla NBs during early pupal development is determined by the exhaustion of the NE stem cell pool. Hence, altering NE-NB transition during larval neurogenesis disrupts the timely termination of medulla NBs. Medulla NBs terminate neurogenesis via a combination of apoptosis, terminal symmetric division via Prospero, and a switch to gliogenesis via Glial Cell Missing (Gcm); however, these processes occur independently of each other. We also show that temporal progression of the medulla NBs is mostly not required for their termination. As the Drosophila OL shares a similar mode of division with mammalian neurogenesis, understanding when and how these progenitors cease proliferation during development can have important implications for mammalian brain size determination and regulation of its overall function.


Every cell in the body can be traced back to a stem cell. For instance, most cells in the adult brains of fruit flies come from a type of stem cell known as a neuroblast. This includes neurons and glial cells (which support and protect neurons) in the optic lobe, the part of the brain that processes visual information. The numbers of neurons and glia in the optic lobe are tightly regulated such that when the right numbers are reached, the neuroblasts stop making more and are terminated. But how and when this occurs is poorly understood. To investigate, Nguyen and Cheng studied when neuroblasts disappear in the optic lobe over the course of development. This revealed that the number of neuroblasts dropped drastically 12 to 18 hours after the fruit fly larvae developed in to pupae, and were completely gone by 30 hours in to pupae life. Further experiments revealed that the timing of this decrease is influenced by neuroepithelium cells, the pool of stem cells that generate neuroblasts during the early stages of development. Nguyen and Cheng found that speeding up this transition so that neuroblasts arise from the neuroepithelium earlier, led neuroblasts to disappear faster from the optic lobe; whereas delaying the transition caused neuroblasts to persist for much longer. Thus, the time at which neuroblasts are born determines when they are terminated. Furthermore, Nguyen and Cheng showed that the neuroblasts were lost through a combination of means. This includes dying via a process called apoptosis, dividing to form two mature neurons, or switching to a glial cell fate. These findings provide a deeper understanding of the mechanisms regulating stem cell pools and their conversion to different cell types, a process that is crucial to the proper development of the brain. How cells divide to form the optic lobe of fruit flies is similar to how new neurons arise in the mammalian brain. Understanding how and when stem cells in the fruit fly brain stop proliferating could therefore provide new insights in to the development of the human brain.


Subject(s)
Apoptosis , Cell Differentiation , Drosophila Proteins , Neural Stem Cells , Neuroepithelial Cells , Neurogenesis , Animals , Neural Stem Cells/physiology , Neural Stem Cells/cytology , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Neurogenesis/physiology , Neuroepithelial Cells/physiology , Neuroepithelial Cells/cytology , Neuroglia/physiology , Neuroglia/cytology , Drosophila/physiology , Drosophila melanogaster/growth & development , Drosophila melanogaster/physiology , Drosophila melanogaster/cytology , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/growth & development , Pupa/growth & development , DNA-Binding Proteins , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL