Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.915
Filter
1.
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
2.
Commun Biol ; 7(1): 1165, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289586

ABSTRACT

EphrinB2 regulates synaptic transmission and morphology however its role in memory formation is unknown. Here we show that deleting ephrinB2 from excitatory neurons in the basolateral amygdala (BLA) of male mice impairs long-term (LTM), but not short-term (STM), fear memory formation. Deleting ephrinB2 from astrocytes in the BLA impairs fear LTM but not STM. Removing ephrinB2 from astrocytes in the BLA reduces the level of the excitatory amino acid transporter 1 (EAAT1) in these cells. Inhibiting EAAT1 activity in the BLA during fear conditioning, by its specific inhibitor UCPH-101, impairs fear LTM showing that EAAT1 in the BLA is needed for fear LTM formation. The administration of ephrinB2 into the BLA during fear conditioning training enhances fear LTM. Moreover, ephrinB2 increases the ability of fear conditioning to activate cells in the BLA as detected by c-Fos labeling. EphrinB2 therefore determines the threshold for fear memory formation. In contrast to mature neurons, we show that ephrinB2 in neural stem cells (NSCs) is not needed for fear LTM. Our study shows that ephrinB2 in the BLA determines the strength of long-term memory consolidation.


Subject(s)
Astrocytes , Basolateral Nuclear Complex , Ephrin-B2 , Fear , Memory, Long-Term , Neurons , Animals , Fear/physiology , Male , Mice , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/physiology , Ephrin-B2/metabolism , Ephrin-B2/genetics , Neurons/metabolism , Neurons/physiology , Astrocytes/metabolism , Astrocytes/physiology , Memory, Long-Term/physiology , Mice, Inbred C57BL
3.
Bull Exp Biol Med ; 177(4): 528-533, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39266920

ABSTRACT

CNS diseases associated with compromised blood supply and/or vascular integrity are one of the leading causes of mortality and disability in adults worldwide and are also among 10 most common causes of death in children. Angiogenesis is an essential element of regeneration processes upon nervous tissue damage and can play a crucial role in neuroprotection. Here we review the features of cerebral vascular regeneration after ischemic stroke, including the complex interactions between endothelial cells and other brain cell types (neural stem cells, astrocytes, microglia, and oligodendrocytes). The mechanisms of reciprocal influence of angiogenesis and neurogenesis, the role of astrocytes in the formation of the blood-brain barrier, and roles of microglia and oligodendrocytes in vascular regeneration are discussed. Understanding the mechanisms of angiogenesis regulation in CNS is of critical importance for the development of new treatments of neurovascular pathologies.


Subject(s)
Astrocytes , Blood-Brain Barrier , Ischemic Stroke , Neovascularization, Physiologic , Neural Stem Cells , Neurogenesis , Humans , Ischemic Stroke/physiopathology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Neovascularization, Physiologic/physiology , Neurogenesis/physiology , Animals , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/physiology , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Oligodendroglia/physiology , Microglia/pathology , Microglia/metabolism , Microglia/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Neuroglia/metabolism , Neuroglia/pathology , Brain Ischemia/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Central Nervous System/blood supply , Central Nervous System/metabolism , Central Nervous System/pathology , Brain/blood supply , Brain/pathology , Brain/metabolism , Brain/physiopathology , Angiogenesis
4.
eNeuro ; 11(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-39227152

ABSTRACT

Astrocytes are essential for the formation and maintenance of neural networks. However, a major technical challenge for investigating astrocyte function and disease-related pathophysiology has been the limited ability to obtain functional human astrocytes. Despite recent advances in human pluripotent stem cell (hPSC) techniques, primary rodent astrocytes remain the gold standard in coculture with human neurons. We demonstrate that a combination of leukemia inhibitory factor (LIF) and bone morphogenetic protein-4 (BMP4) directs hPSC-derived neural precursor cells to a highly pure population of astroglia in 28 d. Using single-cell RNA sequencing, we confirm the astroglial identity of these cells and highlight profound transcriptional adaptations in cocultured hPSC-derived astrocytes and neurons, consistent with their further maturation. In coculture with human neurons, multielectrode array recordings revealed robust network activity of human neurons in a coculture with hPSC-derived or rat astrocytes [3.63 ± 0.44 min-1 (hPSC-derived), 2.86 ± 0.64 min-1 (rat); p = 0.19]. In comparison, we found increased spike frequency within network bursts of human neurons cocultured with hPSC-derived astrocytes [56.31 ± 8.56 Hz (hPSC-derived), 24.77 ± 4.04 Hz (rat); p < 0.01], and whole-cell patch-clamp recordings revealed an increase of postsynaptic currents [2.76 ± 0.39 Hz (hPSC-derived), 1.07 ± 0.14 Hz (rat); p < 0.001], consistent with a corresponding increase in synapse density [14.90 ± 1.27/100 µm2 (hPSC-derived), 8.39 ± 0.63/100 µm2 (rat); p < 0.001]. Taken together, we show that hPSC-derived astrocytes compare favorably with rat astrocytes in supporting human neural network activity and maturation, providing a fully human platform for investigating astrocyte function and neuronal-glial interactions.


Subject(s)
Astrocytes , Coculture Techniques , Neurons , Pluripotent Stem Cells , Astrocytes/physiology , Humans , Animals , Pluripotent Stem Cells/physiology , Rats , Neurons/physiology , Cells, Cultured , Neural Stem Cells/physiology , Cell Differentiation/physiology
5.
Bull Math Biol ; 86(10): 126, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269511

ABSTRACT

During embryonic development of the retina of the eye, astrocytes, a type of glial cell, migrate over the retinal surface and form a dynamic mesh. This mesh then serves as scaffolding for blood vessels to form the retinal vasculature network that supplies oxygen and nutrients to the inner portion of the retina. Astrocyte spreading proceeds in a radially symmetric manner over the retinal surface. Additionally, astrocytes mature from astrocyte precursor cells (APCs) to immature perinatal astrocytes (IPAs) during this embryonic stage. We extend a previously-developed continuum model that describes tension-driven migration and oxygen and growth factor influenced proliferation and differentiation. Comparing numerical simulations to experimental data, we identify model equation components that can be removed via model reduction using approximate Bayesian computation (ABC). Our results verify experimental studies indicating that the choroid oxygen supply plays a negligible role in promoting differentiation of APCs into IPAs and in promoting IPA proliferation, and the hyaloid artery oxygen supply and APC apoptosis play negligible roles in astrocyte spreading and differentiation.


Subject(s)
Astrocytes , Bayes Theorem , Cell Differentiation , Cell Movement , Computer Simulation , Mathematical Concepts , Models, Biological , Retina , Astrocytes/cytology , Astrocytes/physiology , Cell Movement/physiology , Animals , Cell Differentiation/physiology , Retina/cytology , Retina/embryology , Cell Proliferation/physiology , Oxygen/metabolism , Mice
6.
Biol Res ; 57(1): 53, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135103

ABSTRACT

BACKGROUND: As a common disabling disease, irreversible neuronal death due to spinal cord injury (SCI) is the root cause of functional impairment; however, the capacity for neuronal regeneration in the developing spinal cord tissue is limited. Therefore, there is an urgent need to investigate how defective neurons can be replenished and functionally integrated by neural regeneration; the reprogramming of intrinsic cells into functional neurons may represent an ideal solution. METHODS: A mouse model of transection SCI was prepared by forceps clamping, and an adeno-associated virus (AAV) carrying the transcription factors NeuroD1 and Neurogenin-2(Ngn2) was injected in situ into the spinal cord to specifically overexpress these transcription factors in astrocytes close to the injury site. 5-bromo-2´-deoxyuridine (BrdU) was subsequently injected intraperitoneally to continuously track cell regeneration, neuroblasts and immature neurons marker expression, neuronal regeneration, and glial scar regeneration. In addition, immunoprotein blotting was used to measure the levels of transforming growth factor-ß (TGF-ß) pathway-related protein expression. We also evaluated motor function, sensory function, and the integrity of the blood-spinal cord barrier(BSCB). RESULTS: The in situ overexpression of NeuroD1 and Ngn2 in the spinal cord was achieved by specific AAV vectors. This intervention led to a significant increase in cell regeneration and the proportion of cells with neuroblasts and immature neurons cell properties at the injury site(p < 0.0001). Immunofluorescence staining identified astrocytes with neuroblasts and immature neurons cell properties at the site of injury while neuronal marker-specific staining revealed an increased number of mature astrocytes at the injury site. Behavioral assessments showed that the intervention did not improve The BMS (Basso mouse scale) score (p = 0.0726) and gait (p > 0.05), although the treated mice had more sensory sensitivity and greater voluntary motor ability in open field than the non-intervention mice. We observed significant repair of the BSCB at the center of the injury site (p < 0.0001) and a significant improvement in glial scar proliferation. Electrophysiological assessments revealed a significant improvement in spinal nerve conduction (p < 0.0001) while immunostaining revealed that the levels of TGF-ß protein at the site of injury in the intervention group were lower than control group (p = 0.0034); in addition, P70 s6 and PP2A related to the TGF-ß pathway showed ascending trend (p = 0.0036, p = 0.0152 respectively). CONCLUSIONS: The in situ overexpression of NeuroD1 and Ngn2 in the spinal cord after spinal cord injury can reprogram astrocytes into neurons and significantly enhance cell regeneration at the injury site. The reprogramming of astrocytes can lead to tissue repair, thus improving the reduced threshold and increasing voluntary movements. This strategy can also improve the integrity of the blood-spinal cord barrier and enhance nerve conduction function. However, the simple reprogramming of astrocytes cannot lead to significant improvements in the striding function of the lower limbs.


Subject(s)
Astrocytes , Basic Helix-Loop-Helix Transcription Factors , Disease Models, Animal , Nerve Tissue Proteins , Spinal Cord Injuries , Animals , Spinal Cord Injuries/therapy , Spinal Cord Injuries/physiopathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Astrocytes/physiology , Nerve Tissue Proteins/metabolism , Mice , Nerve Regeneration/physiology , Neurons , Female , Mice, Inbred C57BL , Spinal Cord/metabolism
7.
Nat Neurosci ; 27(8): 1444, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107595
8.
Hum Cell ; 37(6): 1638-1648, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39210197

ABSTRACT

Techniques for triggering neural differentiation of embryonic and induced pluripotent stem cells into neural stem cells and neurons have been established. However, neural induction of mesenchymal stem cells, including dental pulp stem cells (DPSCs), has been assessed primarily based on neural-related gene regulation, and detailed studies into the characteristics and differentiation status of cells are lacking. Therefore, this study was aimed at evaluating the cellular components and differentiation pathways of neural lineage cells obtained via neural induction of human DPSCs. Human DPSCs were induced to neural cells in monolayer culture and examined for gene expression and mechanisms underlying differentiation using microarray-based ingenuity pathway analysis. In addition, the neural lineage cells were subjected to single-cell RNA sequencing (scRNA-seq) to classify cell populations based on gene expression profiles and to elucidate their differentiation pathways. Ingenuity pathway analysis revealed that genes exhibiting marked overexpression, post-neuronal induction, such as FABP7 and ZIC1, were associated with neurogenesis. Furthermore, in canonical pathway analysis, axon guidance signals demonstrated maximum activation. The scRNA-seq and cell type annotations revealed the presence of neural progenitor cells, astrocytes, neurons, and a small number of non-neural lineage cells. Moreover, trajectory and pseudotime analyses demonstrated that the neural progenitor cells initially engendered neurons, which subsequently differentiated into astrocytes. This result indicates that the aforementioned neural induction strategy generated neural stem/progenitor cells from DPSCs, which might differentiate and proliferate to constitute neural lineage cells. Therefore, neural induction of DPSCs may present an alternative approach to pluripotent stem cell-based therapeutic interventions for nervous system disorders.


Subject(s)
Cell Differentiation , Dental Pulp , Neural Stem Cells , Neurogenesis , Neurons , Humans , Dental Pulp/cytology , Cell Differentiation/genetics , Neurons/cytology , Neurons/physiology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Cells, Cultured , Neurogenesis/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Gene Expression/genetics , Cell Lineage/genetics , Stem Cells/cytology , Stem Cells/physiology , Astrocytes/cytology , Astrocytes/physiology
9.
Biol Res ; 57(1): 54, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143594

ABSTRACT

Brain damage triggers diverse cellular and molecular events, with astrocytes playing a crucial role in activating local neuroprotective and reparative signaling within damaged neuronal circuits. Here, we investigated reactive astrocytes using a multidimensional approach to categorize their responses into different subtypes based on morphology. This approach utilized the StarTrack lineage tracer, single-cell imaging reconstruction and multivariate data analysis. Our findings identified three profiles of reactive astrocyte responses, categorized by their effects on cell size- and shape- related morphological parameters: "moderate", "strong," and "very strong". We also examined the heterogeneity of astrocyte reactivity, focusing on spatial and clonal distribution. Our research revealed a notable enrichment of protoplasmic and fibrous astrocytes within the "strong" and "very strong" response subtypes. Overall, our study contributes to a better understanding of astrocyte heterogeneity in response to an injury. By characterizing the diverse reactive responses among astrocyte subpopulations, we provide insights that could guide future research aimed at identifying novel therapeutic targets to mitigate brain damage and promote neural repair.


Subject(s)
Astrocytes , Astrocytes/physiology , Animals , Mice , Cell Lineage/physiology , Cluster Analysis , Single-Cell Analysis
10.
Mol Brain ; 17(1): 52, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107815

ABSTRACT

Activation of astrocytes after sensory stimulation has been reported to be involved in increased blood flow in the central nervous system. In the present study, using a chemogenetic method to induce astrocyte activation in mice without sensory stimulation, we found that astrocytic activation led to increased blood flow in the olfactory bulb, suggesting that astrocyte activation is sufficient for increasing blood flow in the olfactory bulb. The technique established here will be useful for studying the mechanisms underlying sensory input-dependent blood flow increases.


Subject(s)
Astrocytes , Olfactory Bulb , Animals , Olfactory Bulb/physiology , Olfactory Bulb/blood supply , Astrocytes/physiology , Mice, Inbred C57BL , Regional Blood Flow/physiology , Male , Mice
11.
Vision Res ; 223: 108461, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39059109

ABSTRACT

Astrocytes serve multiple roles in helping to maintain homeostatic physiology of central nervous system tissue, ranging from metabolic support to coupling between vascular and neural elements. Astrocytes are especially critical in axonal tracts such as the optic nerve, where axons propagate energy-demanding action potentials great distances. In disease, astrocyte remodeling is a dynamic, multifaceted process that is often over-simplified between states of quiescence and reactivity. In glaucoma, axon degeneration in the optic nerve is characterized by progressive stages. So too is astrocyte remodeling. Here, using quantitative analysis of light and electron micrographs of myelinated optic nerve sections from the DBA/2J mouse model of glaucoma, we offer further insight into how astrocyte organization reflects stages of degeneration. This analysis indicates that even as axons degenerate, astrocyte gliosis in the nerve increases without abject proliferation, similar to results in the DBA/2J retina. Gliosis is accompanied by reorganization. As axons expand prior to frank degeneration, astrocyte processes retract from the extra-axonal space and reorient towards the nerve edge. After a critical threshold of expansion, axons drop out, and astrocyte processes distribute more evenly across the nerve reflecting gliosis. This multi-stage process likely reflects local rather than global cues from axons and the surrounding tissue that induce rapid reorganization to promote axon survival and extend functionality of the nerve.


Subject(s)
Astrocytes , Axons , Disease Models, Animal , Glaucoma , Gliosis , Astrocytes/physiology , Astrocytes/pathology , Animals , Glaucoma/physiopathology , Glaucoma/pathology , Mice , Gliosis/pathology , Gliosis/physiopathology , Axons/physiology , Axons/pathology , Mice, Inbred DBA , Optic Nerve/pathology , Hypertrophy
12.
PLoS Biol ; 22(7): e3002712, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38996200

ABSTRACT

Glial cells such as astrocytes can modulate neuronal signaling. Astrocytes can also acquire a reactive phenotype that correlates with cognitive impairments in brain diseases. A study in PLOS Biology shows that prolonged activation of astrocytes can trigger both cognitive impairments and a reactive astrocyte phenotype.


Subject(s)
Astrocytes , Cognition , Astrocytes/physiology , Animals , Humans , Cognition/physiology , Cognitive Dysfunction/physiopathology , Neurons/physiology
13.
J Neurosci ; 44(32)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38955487

ABSTRACT

Recent work demonstrated that activation of spinal D1 and D5 dopamine receptors (D1/D5Rs) facilitates non-Hebbian long-term potentiation (LTP) at primary afferent synapses onto spinal projection neurons. However, the cellular localization of the D1/D5Rs driving non-Hebbian LTP in spinal nociceptive circuits remains unknown, and it is also unclear whether D1/D5R signaling must occur concurrently with sensory input in order to promote non-Hebbian LTP at these synapses. Here we investigate these issues using cell-type-selective knockdown of D1Rs or D5Rs from lamina I spinoparabrachial neurons, dorsal root ganglion (DRG) neurons, or astrocytes in adult mice of either sex using Cre recombinase-based genetic strategies. The LTP evoked by low-frequency stimulation of primary afferents in the presence of the selective D1/D5R agonist SKF82958 persisted following the knockdown of D1R or D5R in spinoparabrachial neurons, suggesting that postsynaptic D1/D5R signaling was dispensable for non-Hebbian plasticity at sensory synapses onto these key output neurons of the superficial dorsal horn (SDH). Similarly, the knockdown of D1Rs or D5Rs in DRG neurons failed to influence SKF82958-enabled LTP in lamina I projection neurons. In contrast, SKF82958-induced LTP was suppressed by the knockdown of D1R or D5R in spinal astrocytes. Furthermore, the data indicate that the activation of D1R/D5Rs in spinal astrocytes can either retroactively or proactively drive non-Hebbian LTP in spinoparabrachial neurons. Collectively, these results suggest that dopaminergic signaling in astrocytes can strongly promote activity-dependent LTP in the SDH, which is predicted to significantly enhance the amplification of ascending nociceptive transmission from the spinal cord to the brain.


Subject(s)
Astrocytes , Long-Term Potentiation , Receptors, Dopamine D1 , Receptors, Dopamine D5 , Synapses , Animals , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/genetics , Long-Term Potentiation/physiology , Astrocytes/metabolism , Astrocytes/physiology , Mice , Male , Receptors, Dopamine D5/metabolism , Receptors, Dopamine D5/agonists , Receptors, Dopamine D5/genetics , Female , Synapses/physiology , Synapses/metabolism , Ganglia, Spinal/cytology , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/cytology , Mice, Transgenic , Mice, Inbred C57BL
14.
Cell Rep ; 43(7): 114504, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38996064

ABSTRACT

Astroglial networks closely interact with neuronal populations, but their functional contribution to neuronal representation of sensory information remains unexplored. The superior colliculus (SC) integrates multi-sensory information by generating distinct spatial patterns of neuronal functional responses to specific sensory stimulation. Here, we report that astrocytes from the mouse SC form extensive networks in the retinorecipient layer compared to visual cortex. This strong astroglial connectivity relies on high expression of gap-junction proteins. Genetic disruption of this connectivity functionally impairs SC retinotopic and orientation preference responses. These alterations are region specific, absent in primary visual cortex, and associated at the circuit level with a specific impairment of collicular neurons synaptic transmission. This has implications for SC-related visually induced innate behavior, as disrupting astroglial networks impairs light-evoked temporary arrest. Our results indicate that astroglial networks shape synaptic circuit activity underlying SC functional visual responses and play a crucial role in integrating visual cues to drive sensory-motor behavior.


Subject(s)
Astrocytes , Neurons , Superior Colliculi , Animals , Astrocytes/metabolism , Astrocytes/physiology , Mice , Superior Colliculi/physiology , Neurons/physiology , Neurons/metabolism , Mice, Inbred C57BL , Synaptic Transmission/physiology , Photic Stimulation , Visual Cortex/physiology , Nerve Net/physiology , Male
15.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-39009447

ABSTRACT

Generation of human induced pluripotent stem cell (hiPSC)-derived motor neurons (MNs) offers an unprecedented approach to modeling movement disorders such as dystonia and amyotrophic lateral sclerosis. However, achieving survival poses a significant challenge when culturing induced MNs, especially when aiming to reach late maturation stages. Utilizing hiPSC-derived motor neurons and primary mouse astrocytes, we assembled two types of coculture systems: direct coculturing of neurons with astrocytes and indirect coculture using culture inserts that physically separate neurons and astrocytes. Both systems significantly enhance neuron survival. Compared with these two systems, no significant differences in neurodevelopment, maturation, and survival within 3 weeks, allowing to prepare neurons at maturation stages. Using the indirect coculture system, we obtained highly pure MNs at the late mature stage from hiPSCs. Transcriptomic studies of hiPSC-derived MNs showed a typical neurodevelopmental switch in gene expression from the early immature stage to late maturation stages. Mature genes associated with neurodevelopment and synaptogenesis are highly enriched in MNs at late stages, demonstrating that these neurons achieve maturation. This study introduces a novel tool for the preparation of highly pure hiPSC-derived neurons, enabling the determination of neurological disease pathogenesis in neurons at late disease onset stages through biochemical approaches, which typically necessitate highly pure neurons. This advancement is particularly significant in modeling age-related neurodegeneration.


Subject(s)
Astrocytes , Coculture Techniques , Induced Pluripotent Stem Cells , Motor Neurons , Induced Pluripotent Stem Cells/physiology , Animals , Motor Neurons/physiology , Mice , Astrocytes/physiology , Humans , Cell Differentiation/physiology , Cells, Cultured , Neurogenesis/physiology
16.
Nat Neurosci ; 27(8): 1475-1488, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39020018

ABSTRACT

Perineuronal nets (PNNs) are densely packed extracellular matrices that cover the cell body of fast-spiking inhibitory neurons. PNNs stabilize synapses inhibiting synaptic plasticity. Here we show that synaptic terminals of fast-spiking interneurons localize to holes in the PNNs in the adult mouse somatosensory cortex. Approximately 95% of holes in the PNNs contain synapses and astrocytic processes expressing Kir4.1, glutamate and GABA transporters. Hence, holes in the PNNs contain tripartite synapses. In the adult mouse brain, PNN degradation causes an expanded astrocytic coverage of the neuronal somata without altering the axon terminals. The loss of PNNs impairs astrocytic transmitter and potassium uptake, resulting in the spillage of glutamate into the extrasynaptic space. Our data show that PNNs and astrocytes cooperate to contain synaptically released signals in physiological conditions. Their combined action is altered in mouse models of Alzheimer's disease and epilepsy where PNNs are disrupted.


Subject(s)
Astrocytes , Extracellular Matrix , Homeostasis , Somatosensory Cortex , Synapses , Animals , Astrocytes/metabolism , Astrocytes/physiology , Mice , Homeostasis/physiology , Synapses/physiology , Synapses/metabolism , Extracellular Matrix/metabolism , Somatosensory Cortex/physiology , Somatosensory Cortex/metabolism , Interneurons/physiology , Interneurons/metabolism , Mice, Inbred C57BL , Male , Mice, Transgenic , Nerve Net/physiology , Neuronal Plasticity/physiology
17.
Exp Neurol ; 379: 114889, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39019303

ABSTRACT

Neuroscience dogma avers that astrocytic "scars" inhibit axonal regeneration after spinal cord injury (SCI). A recent report suggested however that astrocytes form "borders" around lesions that are permissive rather than inhibitory to axonal growth. We now provide further evidence supporting a facilitatory role of astrocytes in axonal regeneration after SCI. First, even 6months after SCI, injured axons are retained within regions of densely reactive astrocytes, in direct contact with astrocyte processes without being repelled. Second, 6 month-delayed implants of neural stem cells extend axons into reactive astrocyte borders surrounding lesions, densely contacting astrocyte surfaces. Third, bioengineered hydrogels implanted into sites of SCI re-orient reactive astrocytic processes to align along the rostral-to-caudal spinal cord axis resulting in successful regeneration into the lesion/scaffold in close association with astrocytic processes. Fourth, corticospinal axons regenerate into neural progenitor cells implanted six months after injury in close association with host astrocytic processes. Thus, astrocytes do not appear to inhibit axonal regeneration, and the close association of newly growing axons with astrocytic processes suggests a facilitatory role in axonal regeneration.


Subject(s)
Astrocytes , Axons , Nerve Regeneration , Spinal Cord Injuries , Spinal Cord Injuries/therapy , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Astrocytes/physiology , Animals , Nerve Regeneration/physiology , Axons/physiology , Rats , Female , Neural Stem Cells/transplantation , Neural Stem Cells/physiology , Disease Models, Animal , Chronic Disease , Rats, Sprague-Dawley
18.
Commun Biol ; 7(1): 852, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997325

ABSTRACT

Astrocytes play a key role in the regulation of synaptic strength and are thought to orchestrate synaptic plasticity and memory. Yet, how specifically astrocytes and their neuroactive transmitters control learning and memory is currently an open question. Recent experiments have uncovered an astrocyte-mediated feedback loop in CA1 pyramidal neurons which is started by the release of endocannabinoids by active neurons and closed by astrocytic regulation of the D-serine levels at the dendrites. D-serine is a co-agonist for the NMDA receptor regulating the strength and direction of synaptic plasticity. Activity-dependent D-serine release mediated by astrocytes is therefore a candidate for mediating between long-term synaptic depression (LTD) and potentiation (LTP) during learning. Here, we show that the mathematical description of this mechanism leads to a biophysical model of synaptic plasticity consistent with the phenomenological model known as the BCM model. The resulting mathematical framework can explain the learning deficit observed in mice upon disruption of the D-serine regulatory mechanism. It shows that D-serine enhances plasticity during reversal learning, ensuring fast responses to changes in the external environment. The model provides new testable predictions about the learning process, driving our understanding of the functional role of neuron-glia interaction in learning.


Subject(s)
Astrocytes , Neuronal Plasticity , Reversal Learning , Animals , Astrocytes/physiology , Astrocytes/metabolism , Neuronal Plasticity/physiology , Mice , Reversal Learning/physiology , Serine/metabolism , Models, Neurological , Receptors, N-Methyl-D-Aspartate/metabolism
19.
Proc Natl Acad Sci U S A ; 121(28): e2317711121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968101

ABSTRACT

Adult neural stem cells (NSCs) reside in the dentate gyrus of the hippocampus, and their capacity to generate neurons and glia plays a role in learning and memory. In addition, neurodegenerative diseases are known to be caused by a loss of neurons and glial cells, resulting in a need to better understand stem cell fate commitment processes. We previously showed that NSC fate commitment toward a neuronal or glial lineage is strongly influenced by extracellular matrix stiffness, a property of elastic materials. However, tissues in vivo are not purely elastic and have varying degrees of viscous character. Relatively little is known about how the viscoelastic properties of the substrate impact NSC fate commitment. Here, we introduce a polyacrylamide-based cell culture platform that incorporates mismatched DNA oligonucleotide-based cross-links as well as covalent cross-links. This platform allows for tunable viscous stress relaxation properties via variation in the number of mismatched base pairs. We find that NSCs exhibit increased astrocytic differentiation as the degree of stress relaxation is increased. Furthermore, culturing NSCs on increasingly stress-relaxing substrates impacts cytoskeletal dynamics by decreasing intracellular actin flow rates and stimulating cyclic activation of the mechanosensitive protein RhoA. Additionally, inhibition of motor-clutch model components such as myosin II and focal adhesion kinase partially or completely reverts cells to lineage distributions observed on elastic substrates. Collectively, our results introduce a unique system for controlling matrix stress relaxation properties and offer insight into how NSCs integrate viscoelastic cues to direct fate commitment.


Subject(s)
Cell Differentiation , Neural Stem Cells , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/physiology , Mice , Acrylic Resins/chemistry , rhoA GTP-Binding Protein/metabolism , Cells, Cultured , Neurons/metabolism , Neurons/physiology , Neurons/cytology , Extracellular Matrix/metabolism , Stress, Mechanical
20.
Mol Cell Neurosci ; 130: 103947, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38862082

ABSTRACT

As the main players in the central nervous system (CNS), neurons dominate most life activities. However, after accidental trauma or neurodegenerative diseases, neurons are unable to regenerate themselves. The loss of this important role can seriously affect the quality of life of patients, ranging from movement disorders to disability and even death. There is no suitable treatment to prevent or reverse this process. Therefore, the regeneration of neurons after loss has been a major clinical problem and the key to treatment. Replacing the lost neurons by transdifferentiation of other cells is the only viable approach. Although much progress has been made in stem cell therapy, ethical issues, immune rejection, and limited cell sources still hinder its clinical application. In recent years, somatic cell reprogramming technology has brought a new dawn. Among them, astrocytes, as endogenously abundant cells homologous to neurons, have good potential and application value for reprogramming into neurons, having been reprogrammed into neurons in vitro and in vivo in a variety of ways.


Subject(s)
Astrocytes , Cellular Reprogramming , Neurons , Humans , Astrocytes/metabolism , Astrocytes/physiology , Astrocytes/cytology , Animals , Neurons/physiology , Neurons/metabolism , Neurons/cytology , Cellular Reprogramming/physiology , Cell Transdifferentiation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL