Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.879
Filter
1.
Int J Neural Syst ; 34(6): 2450028, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38706265

ABSTRACT

Spiking neural membrane systems (or spiking neural P systems, SNP systems) are a new type of computation model which have attracted the attention of plentiful scholars for parallelism, time encoding, interpretability and extensibility. The original SNP systems only consider the time delay caused by the execution of rules within neurons, but not caused by the transmission of spikes via synapses between neurons and its adaptive adjustment. In view of the importance of time delay for SNP systems, which are a time encoding computation model, this study proposes SNP systems with adaptive synaptic time delay (ADSNP systems) based on the dynamic regulation mechanism of synaptic transmission delay in neural systems. In ADSNP systems, besides neurons, astrocytes that can generate adenosine triphosphate (ATP) are introduced. After receiving spikes, astrocytes convert spikes into ATP and send ATP to the synapses controlled by them to change the synaptic time delays. The Turing universality of ADSNP systems in number generating and accepting modes is proved. In addition, a small universal ADSNP system using 93 neurons and astrocytes is given. The superiority of the ADSNP system is demonstrated by comparison with the six variants. Finally, an ADSNP system is constructed for credit card fraud detection, which verifies the feasibility of the ADSNP system for solving real-world problems. By considering the adaptive synaptic delay, ADSNP systems better restore the process of information transmission in biological neural networks, and enhance the adaptability of SNP systems, making the control of time more accurate.


Subject(s)
Astrocytes , Models, Neurological , Neural Networks, Computer , Neurons , Synapses , Synaptic Transmission , Synapses/physiology , Astrocytes/physiology , Neurons/physiology , Synaptic Transmission/physiology , Action Potentials/physiology , Adenosine Triphosphate/metabolism , Time Factors , Humans
2.
J Neurosci Res ; 102(5): e25356, 2024 May.
Article in English | MEDLINE | ID: mdl-38773875

ABSTRACT

From the blood brain barrier to the synaptic space, astrocytes provide structural, metabolic, ionic, and extracellular matrix (ECM) support across the brain. Astrocytes include a vast array of subtypes, their phenotypes and functions varying both regionally and temporally. Astrocytes' metabolic and regulatory functions poise them to be quick and sensitive responders to injury and disease in the brain as revealed by single cell sequencing. Far less is known about the influence of the local healthy and aging microenvironments on these astrocyte activation states. In this forward-looking review, we describe the known relationship between astrocytes and their local microenvironment, the remodeling of the microenvironment during disease and injury, and postulate how they may drive astrocyte activation. We suggest technology development to better understand the dynamic diversity of astrocyte activation states, and how basal and activation states depend on the ECM microenvironment. A deeper understanding of astrocyte response to stimuli in ECM-specific contexts (brain region, age, and sex of individual), paves the way to revolutionize how the field considers astrocyte-ECM interactions in brain injury and disease and opens routes to return astrocytes to a healthy quiescent state.


Subject(s)
Astrocytes , Brain , Extracellular Matrix , Astrocytes/physiology , Astrocytes/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Humans , Animals , Brain/metabolism , Brain Injuries/pathology , Brain Injuries/metabolism
3.
Sci Signal ; 17(838): eadq5728, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38805584
4.
BMC Biol ; 22(1): 75, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38566045

ABSTRACT

BACKGROUND: Trans-differentiation of human-induced pluripotent stem cells into neurons via Ngn2-induction (hiPSC-N) has become an efficient system to quickly generate neurons a likely significant advance for disease modeling and in vitro assay development. Recent single-cell interrogation of Ngn2-induced neurons, however, has revealed some similarities to unexpected neuronal lineages. Similarly, a straightforward method to generate hiPSC-derived astrocytes (hiPSC-A) for the study of neuropsychiatric disorders has also been described. RESULTS: Here, we examine the homogeneity and similarity of hiPSC-N and hiPSC-A to their in vivo counterparts, the impact of different lengths of time post Ngn2 induction on hiPSC-N (15 or 21 days), and the impact of hiPSC-N/hiPSC-A co-culture. Leveraging the wealth of existing public single-cell RNA-seq (scRNA-seq) data in Ngn2-induced neurons and in vivo data from the developing brain, we provide perspectives on the lineage origins and maturation of hiPSC-N and hiPSC-A. While induction protocols in different labs produce consistent cell type profiles, both hiPSC-N and hiPSC-A show significant heterogeneity and similarity to multiple in vivo cell fates, and both more precisely approximate their in vivo counterparts when co-cultured. Gene expression data from the hiPSC-N show enrichment of genes linked to schizophrenia (SZ) and autism spectrum disorders (ASD) as has been previously shown for neural stem cells and neurons. These overrepresentations of disease genes are strongest in our system at early times (day 15) in Ngn2-induction/maturation of neurons, when we also observe the greatest similarity to early in vivo excitatory neurons. We have assembled this new scRNA-seq data along with the public data explored here as an integrated biologist-friendly web-resource for researchers seeking to understand this system more deeply: https://nemoanalytics.org/p?l=DasEtAlNGN2&g=NES . CONCLUSIONS: While overall we support the use of the investigated cellular models for the study of neuropsychiatric disease, we also identify important limitations. We hope that this work will contribute to understanding and optimizing cellular modeling for complex brain disorders.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Coculture Techniques , Astrocytes/physiology , Neurons/physiology , Cell Differentiation , Gene Expression Profiling
5.
Medicina (Kaunas) ; 60(4)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38674304

ABSTRACT

Background and Objectives. Neurogenesis is an integral process in post-stroke recovery, involving the recruitment of proliferating neuroblasts from neurogenic niches of the mammal brain. However, the role of neurogenesis in the long-term restoration following ischemic stroke is fragmented. Post-stroke motor dysfunction includes challenges in the proper, coordinated use of hands and is present in roughly two-thirds of human patients. In this study, we investigated chronic behavioral and biochemical alterations after transient cerebral ischemia in adult male mice. Materials and Methods: Twelve-week-old C57BL/6N male mice were used, and fMCAo lasting 60 min was induced. At multiple timepoints after fMCAo induction, a single pellet reaching task was performed. Six months after the procedure, we immunohistochemically determined the number of proliferating neuroblasts (BrdU and DCX-positive) and the number of differentiated astrocytes (GFAP-positive) in both brain hemispheres. Results: The reaching ability of fMCAo mice was impaired from one month to six months after the induction of ischemia. Neuroblast proliferation was increased in the ipsilateral SVZ, whereas GFAP+ cell count was elevated in the hippocampal DG of both hemispheres of the fMCAo group mice. Conclusions: Our current report demonstrates the long-term effects of transient cerebral ischemia on mice functional parameters and neurogenesis progression. Our data demonstrate that transient cerebral ischemia promotes a long-lasting regenerative response in the ipsilateral brain hemisphere, specifically in the neurogenic SVZ and DG regions.


Subject(s)
Astrocytes , Doublecortin Protein , Mice, Inbred C57BL , Motor Skills , Neurogenesis , Animals , Neurogenesis/physiology , Mice , Male , Astrocytes/physiology , Motor Skills/physiology , Disease Models, Animal , Ischemic Attack, Transient/physiopathology , Ischemic Attack, Transient/complications
6.
Glia ; 72(7): 1356-1370, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38591270

ABSTRACT

Astrocytes throughout the central nervous system are heterogeneous in both structure and function. This diversity leads to tissue-specific specialization where morphology is adapted to the surrounding neuronal circuitry, as seen in Bergman glia of the cerebellum and Müller glia of the retina. Because morphology can be a differentiating factor for cellular classification, we recently developed a mouse where glial-fibrillary acidic protein (GFAP)-expressing cells stochastically label for full membranous morphology. Here we utilize this tool to investigate whether morphological and electrophysiological features separate types of mouse retinal astrocytes. In this work, we report on a novel glial population found in the inner plexiform layer and ganglion cell layer which expresses the canonical astrocyte markers GFAP, S100ß, connexin-43, Sox2 and Sox9. Apart from their retinal layer localization, these cells are unique in their radial distribution. They are notably absent from the mid-retina but are heavily concentrated near the optic nerve head, and to a lesser degree the peripheral retina. Additionally, their morphology is distinct from both nerve fiber layer astrocytes and Müller glia, appearing more similar to amacrine cells. Despite this structural similarity, these cells lack protein expression of common neuronal markers. Additionally, they do not exhibit action potentials, but rather resemble astrocytes and Müller glia in their small amplitude, graded depolarization to both light onset and offset. Their structure, protein expression, physiology, and intercellular connections suggest that these cells are astrocytic, displaced from their counterparts in the nerve fiber layer. As such, we refer to these cells as displaced retinal astrocytes.


Subject(s)
Astrocytes , Mice, Transgenic , Retina , Animals , Astrocytes/metabolism , Astrocytes/physiology , Retina/cytology , Retina/metabolism , Retina/physiology , Mice , Glial Fibrillary Acidic Protein/metabolism , Mice, Inbred C57BL , Action Potentials/physiology
7.
J Neurosci Methods ; 407: 110127, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38615721

ABSTRACT

BACKGROUND: Human induced pluripotent stem cell (hiPSC)- derived neurons offer the possibility of studying human-specific neuronal behaviors in physiologic and pathologic states in vitro. It is unclear whether cultured neurons can achieve the fundamental network behaviors required to process information in the brain. Investigating neuronal oscillations and their interactions, as occurs in cross-frequency coupling (CFC), addresses this question. NEW METHODS: We examined whether networks of two-dimensional (2D) cultured hiPSC-derived cortical neurons grown with hiPSC-derived astrocytes on microelectrode array plates recapitulate the CFC that is present in vivo. We employed the modulation index method for detecting phase-amplitude coupling (PAC) and used offline spike sorting to analyze the contribution of single neuron spiking to network behavior. RESULTS: We found that PAC is present, the degree of PAC is specific to network structure, and it is modulated by external stimulation with bicuculline administration. Modulation of PAC is not driven by single neurons, but by network-level interactions. COMPARISON WITH EXISTING METHODS: PAC has been demonstrated in multiple regions of the human cortex as well as in organoids. This is the first report of analysis demonstrating the presence of coupling in 2D cultures. CONCLUSION: CFC in the form of PAC analysis explores communication and integration between groups of neurons and dynamical changes across networks. In vitro PAC analysis has the potential to elucidate the underlying mechanisms as well as capture the effects of chemical, electrical, or ultrasound stimulation; providing insight into modulation of neural networks to treat nervous system disorders in vivo.


Subject(s)
Induced Pluripotent Stem Cells , Microelectrodes , Neurons , Humans , Neurons/physiology , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/cytology , Action Potentials/physiology , Cells, Cultured , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Astrocytes/physiology , Cell Culture Techniques/methods , Cell Culture Techniques/instrumentation , Bicuculline/pharmacology , Nerve Net/physiology
8.
Behav Brain Res ; 468: 115017, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38679145

ABSTRACT

Growing evidence indicates a critical role of astrocytes in learning and memory. However, little is known about the role of basolateral amygdala complex (BLA-C) astrocytes in contextual fear conditioning (CFC), a paradigm relevant to understand and generate treatments for fear- and anxiety-related disorders. To get insights on the involvement of BLA-C astrocytes in fear memory, fluorocitrate (FLC), a reversible astroglial metabolic inhibitor, was applied at critical moments of the memory processing in order to target the acquisition, consolidation, retrieval and reconsolidation process of the fear memory. Adult Wistar male rats were bilaterally cannulated in BLA-C. Ten days later they were infused with different doses of FLC (0.5 or 1 nmol/0.5 µl) or saline before or after CFC and before or after retrieval. FLC impaired fear memory expression when administered before and shortly after CFC, but not one hour later. Infusion of FLC prior and after retrieval did not affect the memory. Our findings suggest that BLA-C astrocytes are critically involved in the acquisition/early consolidation of fear memory but not in the retrieval and reconsolidation. Furthermore, the extinction process was presumably not affected (considering that peri-retrieval administration could also affect this process).


Subject(s)
Astrocytes , Basolateral Nuclear Complex , Fear , Memory , Rats, Wistar , Animals , Fear/physiology , Fear/drug effects , Astrocytes/drug effects , Astrocytes/physiology , Male , Basolateral Nuclear Complex/drug effects , Basolateral Nuclear Complex/physiology , Rats , Memory/physiology , Memory/drug effects , Citrates/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Memory Consolidation/physiology , Memory Consolidation/drug effects , Amygdala/drug effects , Amygdala/physiology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology
9.
Nat Neurosci ; 27(5): 927-939, 2024 May.
Article in English | MEDLINE | ID: mdl-38570661

ABSTRACT

An essential feature of neurons is their ability to centrally integrate information from their dendrites. The activity of astrocytes, in contrast, has been described as mostly uncoordinated across cellular compartments without clear central integration. Here we report conditional integration of calcium signals in astrocytic distal processes at their soma. In the hippocampus of adult mice of both sexes, we found that global astrocytic activity, as recorded with population calcium imaging, reflected past neuronal and behavioral events on a timescale of seconds. Salient past events, indicated by pupil dilations, facilitated the propagation of calcium signals from distal processes to the soma. Centripetal propagation to the soma was reproduced by optogenetic activation of the locus coeruleus, a key regulator of arousal, and reduced by pharmacological inhibition of α1-adrenergic receptors. Together, our results suggest that astrocytes are computational units of the brain that slowly and conditionally integrate calcium signals upon behaviorally relevant events.


Subject(s)
Astrocytes , Calcium Signaling , Hippocampus , Locus Coeruleus , Animals , Locus Coeruleus/physiology , Locus Coeruleus/cytology , Astrocytes/physiology , Mice , Hippocampus/physiology , Hippocampus/cytology , Male , Calcium Signaling/physiology , Female , Optogenetics , Mice, Transgenic , Neurons/physiology , Mice, Inbred C57BL , Calcium/metabolism
11.
Cells ; 13(7)2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38607045

ABSTRACT

In vitro and preclinical in vivo research in the last 35 years has clearly highlighted the crucial physiopathological role of glial cells, namely astrocytes/microglia/oligodendrocytes and satellite glial cells/Schwann cells in the central and peripheral nervous system, respectively. Several possible pharmacological targets to various neurodegenerative disorders and painful conditions have therefore been successfully identified, including receptors and enzymes, and mediators of neuroinflammation. However, the translation of these promising data to a clinical setting is often hampered by both technical and biological difficulties, making it necessary to perform experiments on human cells and models of the various diseases. In this review we will, therefore, summarize the most relevant data on the contribution of glial cells to human pathologies and on their possible pharmacological modulation based on data obtained in post-mortem tissues and in iPSC-derived human brain cells and organoids. The possibility of an in vivo visualization of glia reaction to neuroinflammation in patients will be also discussed.


Subject(s)
Neuroglia , Neuroinflammatory Diseases , Humans , Central Nervous System , Microglia/physiology , Astrocytes/physiology
12.
Front Biosci (Landmark Ed) ; 29(3): 94, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38538273

ABSTRACT

Activation of astrocytes during spinal cord injury (SCI) is accompanied by changes in their morphology and functional activity, possibly having severity-, localization-, and time-dependent features. The understanding of the role of reactive astrocytes has undergone significant changes over the last decades, and new data are still emerging to assess the diversity of functional manifestations of reactive cells. This review discusses the current understanding of astrocyte behavior, possible manifestations of their negative and positive roles in SCI, and the prospects for using various methods of directed polarization of astrocytes to improve post-traumatic outcomes. Despite the existing difficulties regarding the disclosure of the complex cascade of molecular changes of reactive astrocytes in different posttraumatic periods, researchers do not give up hope for the development of astrocyte-targeted methods that could reduce the severity of secondary injury by regulating the negative effects of these cells.


Subject(s)
Astrocytes , Spinal Cord Injuries , Animals , Humans , Astrocytes/physiology , Disease Models, Animal , Spinal Cord
13.
Biomolecules ; 14(3)2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38540709

ABSTRACT

Today, neurodegenerative disorders like Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) affect millions of people worldwide, and as the average human lifespan increases, similarly grows the number of patients. For many decades, cognitive and motoric decline has been explained by the very apparent deterioration of neurons in various regions of the brain and spinal cord. However, more recent studies show that disease progression is greatly influenced by the vast population of glial cells. Astrocytes are traditionally considered star-shaped cells on which neurons rely heavily for their optimal homeostasis and survival. Increasing amounts of evidence depict how astrocytes lose their supportive functions while simultaneously gaining toxic properties during neurodegeneration. Many of these changes are similar across various neurodegenerative diseases, and in this review, we highlight these commonalities. We discuss how astrocyte dysfunction drives neuronal demise across a wide range of neurodegenerative diseases, but rather than categorizing based on disease, we aim to provide an overview based on currently known mechanisms. As such, this review delivers a different perspective on the disease causes of neurodegeneration in the hope to encourage further cross-disease studies into shared disease mechanisms, which might ultimately disclose potentially common therapeutic entry points across a wide panel of neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Parkinson Disease , Humans , Astrocytes/physiology , Amyotrophic Lateral Sclerosis/therapy
14.
Trends Neurosci ; 47(4): 289-302, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38521710

ABSTRACT

Selective vulnerability of specific brain regions and cell populations is a hallmark of neurodegenerative disorders. Mechanisms of selective vulnerability involve neuronal heterogeneity, functional specializations, and differential sensitivities to stressors and pathogenic factors. In this review we discuss the growing body of literature suggesting that, like neurons, astrocytes are heterogeneous and specialized, respond to and integrate diverse inputs, and induce selective effects on brain function. In disease, astrocytes undergo specific, context-dependent changes that promote different pathogenic trajectories and functional outcomes. We propose that astrocytes contribute to selective vulnerability through maladaptive transitions to context-divergent phenotypes that impair specific brain regions and functions. Further studies on the multifaceted roles of astrocytes in disease may provide new therapeutic approaches to enhance resilience against neurodegenerative disorders.


Subject(s)
Astrocytes , Neurodegenerative Diseases , Humans , Astrocytes/physiology , Neurodegenerative Diseases/pathology , Neurons/physiology , Brain/pathology
15.
Commun Biol ; 7(1): 373, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38548965

ABSTRACT

Astrocytes in the retrotrapezoid nucleus (RTN) stimulate breathing in response to CO2/H+, however, it is not clear how these cells detect changes in CO2/H+. Considering Kir4.1/5.1 channels are CO2/H+-sensitive and important for several astrocyte-dependent processes, we consider Kir4.1/5.1 a leading candidate CO2/H+ sensor in RTN astrocytes. To address this, we show that RTN astrocytes express Kir4.1 and Kir5.1 transcripts. We also characterized respiratory function in astrocyte-specific inducible Kir4.1 knockout mice (Kir4.1 cKO); these mice breathe normally under room air conditions but show a blunted ventilatory response to high levels of CO2, which could be partly rescued by viral mediated re-expression of Kir4.1 in RTN astrocytes. At the cellular level, astrocytes in slices from astrocyte-specific inducible Kir4.1 knockout mice are less responsive to CO2/H+ and show a diminished capacity for paracrine modulation of respiratory neurons. These results suggest Kir4.1/5.1 channels in RTN astrocytes contribute to respiratory behavior.


Subject(s)
Astrocytes , Carbon Dioxide , Mice , Animals , Astrocytes/physiology , Respiration , Neurons/physiology , Mice, Knockout
16.
Physiology (Bethesda) ; 39(4): 0, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38530221

ABSTRACT

Astrocytes are no longer considered as passive support cells. In the hypothalamus, these glial cells actively participate in the control of appetite, energy expenditure, and the processes leading to obesity and its secondary complications. Here we briefly review studies supporting this conclusion and the advances made in understanding the underlying mechanisms.


Subject(s)
Astrocytes , Energy Metabolism , Hypothalamus , Neurons , Astrocytes/metabolism , Astrocytes/physiology , Hypothalamus/metabolism , Hypothalamus/physiology , Animals , Humans , Neurons/physiology , Neurons/metabolism , Energy Metabolism/physiology , Obesity/metabolism , Obesity/physiopathology
17.
J Cereb Blood Flow Metab ; 44(6): 881-895, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38513138

ABSTRACT

The blood-brain barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional changes during aging, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. In recent years, advances in microscopy and high-throughput bioinformatics have allowed a more in-depth investigation of the aging mechanisms of BBB. This review summarizes age-related alterations of the BBB structure and function from six perspectives: endothelial cells, astrocytes, pericytes, basement membrane, microglia and perivascular macrophages, and fibroblasts, ranging from the molecular level to the human multi-system level. These basic components are essential for the proper functioning of the BBB. Recent imaging methods of BBB were also reviewed. Elucidation of age-associated BBB changes may offer insights into BBB homeostasis and may provide effective therapeutic strategies to protect it during aging.


Subject(s)
Aging , Blood-Brain Barrier , Blood-Brain Barrier/metabolism , Humans , Aging/physiology , Animals , Endothelial Cells/metabolism , Pericytes/metabolism , Pericytes/physiology , Astrocytes/metabolism , Astrocytes/physiology
18.
Glia ; 72(7): 1290-1303, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38506330

ABSTRACT

Astrocytes represent a diverse and morphologically complex group of glial cells critical for shaping and maintaining nervous system homeostasis, as well as responding to injuries. Understanding the origins of astroglial heterogeneity, originated from a limited number of progenitors, has been the focus of many studies. Most of these investigations have centered on protoplasmic and pial astrocytes, while the clonal relationship of fibrous astrocytes or juxtavascular astrocytes has remained relatively unexplored. In this study, we sought to elucidate the morphological diversity and clonal distribution of astrocytes across adult cortical layers, with particular emphasis on their ontogenetic origins. Using the StarTrack lineage tracing tool, we explored the characteristics of adult astroglial clones derived from single and specific progenitors at various embryonic stages. Our results revealed a heterogeneous spatial distribution of astroglial clones, characterized by variations in location, clonal size, and rostro-caudal dispersion. While a considerable proportion of clones were confined within specific cortical layers, others displayed sibling cells crossing layer boundaries. Notably, we observed a correlation between clone location and developmental stage at earlier embryonic stages, although this relationship diminished in later stages. Fibrous astrocyte clones were exclusively confined to the corpus callosum. In contrast, protoplasmic or juxtavascular clones were located in either the upper or lower cortical layers, with certain clones displayed sibling cells distributed across both regions. Our findings underscore the developmental origins and spatial distribution of astroglial clones within cortical layers, providing new insights into the interplay between their morphology, clonal sizes, and progenitor heterogeneity.


Subject(s)
Astrocytes , Astrocytes/cytology , Astrocytes/physiology , Animals , Clone Cells , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Cortex/embryology , Mice, Transgenic , Mice , Neural Stem Cells/cytology , Neural Stem Cells/physiology
19.
Nature ; 627(8003): 374-381, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38326616

ABSTRACT

Memory encodes past experiences, thereby enabling future plans. The basolateral amygdala is a centre of salience networks that underlie emotional experiences and thus has a key role in long-term fear memory formation1. Here we used spatial and single-cell transcriptomics to illuminate the cellular and molecular architecture of the role of the basolateral amygdala in long-term memory. We identified transcriptional signatures in subpopulations of neurons and astrocytes that were memory-specific and persisted for weeks. These transcriptional signatures implicate neuropeptide and BDNF signalling, MAPK and CREB activation, ubiquitination pathways, and synaptic connectivity as key components of long-term memory. Notably, upon long-term memory formation, a neuronal subpopulation defined by increased Penk and decreased Tac expression constituted the most prominent component of the memory engram of the basolateral amygdala. These transcriptional changes were observed both with single-cell RNA sequencing and with single-molecule spatial transcriptomics in intact slices, thereby providing a rich spatial map of a memory engram. The spatial data enabled us to determine that this neuronal subpopulation interacts with adjacent astrocytes, and functional experiments show that neurons require interactions with astrocytes to encode long-term memory.


Subject(s)
Astrocytes , Cell Communication , Gene Expression Profiling , Memory, Long-Term , Neurons , Astrocytes/cytology , Astrocytes/metabolism , Astrocytes/physiology , Basolateral Nuclear Complex/cytology , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/physiology , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Memory, Long-Term/physiology , Mitogen-Activated Protein Kinases/metabolism , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Sequence Analysis, RNA , Single Molecule Imaging , Single-Cell Gene Expression Analysis , Ubiquitination
20.
Cells ; 13(4)2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38391931

ABSTRACT

Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system (CNS). Most GABAergic neurons synthesize GABA from glutamate and release it in the synaptic cleft in the CNS. However, astrocytes can also synthesize and release GABA, activating GABA receptors in the neighboring neurons in physiological and pathological conditions. As the primary homeostatic glial cells in the brain, astrocytes play a crucial role in regulating GABA homeostasis and synaptic neurotransmission. Accumulating evidence demonstrates that astrocytic GABA dysregulation is implicated in psychiatric disorders, including alcohol use disorder (AUD) and major depressive disorder (MDD), the most prevalent co-occurring psychiatric disorders. Several current medications and emerging pharmacological agents targeting GABA levels are in clinical trials for treating AUD and MDD. This review offers a concise summary of the role of astrocytic GABA regulation in AUD and MDD. We also provide an overview of the current understanding and areas of debate regarding the mechanisms by which astrocytes regulate GABA in the CNS and their potential significance in the molecular basis of AUD and MDD, paving the way toward future research directions and potential therapeutic target areas within this field.


Subject(s)
Depressive Disorder, Major , Humans , Astrocytes/physiology , gamma-Aminobutyric Acid , Receptors, GABA , GABAergic Neurons
SELECTION OF CITATIONS
SEARCH DETAIL
...