Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 14.664
1.
Proc Natl Acad Sci U S A ; 121(20): e2321711121, 2024 May 14.
Article En | MEDLINE | ID: mdl-38713624

During development, neural stem cells in the cerebral cortex, also known as radial glial cells (RGCs), generate excitatory neurons, followed by production of cortical macroglia and inhibitory neurons that migrate to the olfactory bulb (OB). Understanding the mechanisms for this lineage switch is fundamental for unraveling how proper numbers of diverse neuronal and glial cell types are controlled. We and others recently showed that Sonic Hedgehog (Shh) signaling promotes the cortical RGC lineage switch to generate cortical oligodendrocytes and OB interneurons. During this process, cortical RGCs generate intermediate progenitor cells that express critical gliogenesis genes Ascl1, Egfr, and Olig2. The increased Ascl1 expression and appearance of Egfr+ and Olig2+ cortical progenitors are concurrent with the switch from excitatory neurogenesis to gliogenesis and OB interneuron neurogenesis in the cortex. While Shh signaling promotes Olig2 expression in the developing spinal cord, the exact mechanism for this transcriptional regulation is not known. Furthermore, the transcriptional regulation of Olig2 and Egfr has not been explored. Here, we show that in cortical progenitor cells, multiple regulatory programs, including Pax6 and Gli3, prevent precocious expression of Olig2, a gene essential for production of cortical oligodendrocytes and astrocytes. We identify multiple enhancers that control Olig2 expression in cortical progenitors and show that the mechanisms for regulating Olig2 expression are conserved between the mouse and human. Our study reveals evolutionarily conserved regulatory logic controlling the lineage switch of cortical neural stem cells.


Basic Helix-Loop-Helix Transcription Factors , Cerebral Cortex , ErbB Receptors , Hedgehog Proteins , Nerve Tissue Proteins , Neural Stem Cells , Neurogenesis , Oligodendrocyte Transcription Factor 2 , PAX6 Transcription Factor , Animals , Neurogenesis/physiology , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Mice , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendrocyte Transcription Factor 2/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , PAX6 Transcription Factor/metabolism , PAX6 Transcription Factor/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics , Neuroglia/metabolism , Neuroglia/cytology , Gene Expression Regulation, Developmental , Signal Transduction , Olfactory Bulb/metabolism , Olfactory Bulb/cytology , Cell Lineage , Humans
2.
Science ; 384(6696): eadk4858, 2024 May 10.
Article En | MEDLINE | ID: mdl-38723085

To fully understand how the human brain works, knowledge of its structure at high resolution is needed. Presented here is a computationally intensive reconstruction of the ultrastructure of a cubic millimeter of human temporal cortex that was surgically removed to gain access to an underlying epileptic focus. It contains about 57,000 cells, about 230 millimeters of blood vessels, and about 150 million synapses and comprises 1.4 petabytes. Our analysis showed that glia outnumber neurons 2:1, oligodendrocytes were the most common cell, deep layer excitatory neurons could be classified on the basis of dendritic orientation, and among thousands of weak connections to each neuron, there exist rare powerful axonal inputs of up to 50 synapses. Further studies using this resource may bring valuable insights into the mysteries of the human brain.


Neurons , Synapses , Temporal Lobe , Humans , Neurons/ultrastructure , Synapses/physiology , Synapses/ultrastructure , Oligodendroglia/cytology , Neuroglia , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/ultrastructure , Dendrites/physiology , Axons/physiology , Axons/ultrastructure
3.
Neuron ; 112(9): 1373-1375, 2024 May 01.
Article En | MEDLINE | ID: mdl-38697018

Maternal well-being is important for the development of the fetus, with a key influence on its nervous system. In this issue of Neuron, Krontira et al.1 implicate glucocorticoids, the stress hormones, in the regulation of neural stem cell identity and proliferation, with long-lasting consequences on brain architecture and educational attainment.


Glucocorticoids , Neurogenesis , Humans , Glucocorticoids/pharmacology , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/cytology , Neural Stem Cells/drug effects
4.
Trends Immunol ; 45(5): 327-328, 2024 May.
Article En | MEDLINE | ID: mdl-38664101

Lawrence et al. report that fetal cortical boundaries are susceptible to morphogenetic stress that regulates a microglia state resembling postnatal, axon-tract associated microglia (ATM). This state performs a newfound function at these boundaries by preventing the formation of cavitary lesions, mediated in part by Spp1-regulated phagocytosis of fibronectin 1.


Microglia , Microglia/physiology , Animals , Humans , Phagocytosis , Cerebral Cortex/embryology , Cerebral Cortex/cytology , Brain/embryology , Brain/physiology , Fibronectins/metabolism
5.
EMBO Rep ; 25(5): 2202-2219, 2024 May.
Article En | MEDLINE | ID: mdl-38600346

Neural progenitor cells within the cerebral cortex undergo a characteristic switch between symmetric self-renewing cell divisions early in development and asymmetric neurogenic divisions later. Yet, the mechanisms controlling this transition remain unclear. Previous work has shown that early but not late neural progenitor cells (NPCs) endogenously express the autism-linked transcription factor Foxp1, and both loss and gain of Foxp1 function can alter NPC activity and fate choices. Here, we show that premature loss of Foxp1 upregulates transcriptional programs regulating angiogenesis, glycolysis, and cellular responses to hypoxia. These changes coincide with a premature destabilization of HIF-1α, an elevation in HIF-1α target genes, including Vegfa in NPCs, and precocious vascular network development. In vitro experiments demonstrate that stabilization of HIF-1α in Foxp1-deficient NPCs rescues the premature differentiation phenotype and restores NPC maintenance. Our data indicate that the endogenous decline in Foxp1 expression activates the HIF-1α transcriptional program leading to changes in the tissue environment adjacent to NPCs, which, in turn, might alter their self-renewal and neurogenic capacities.


Cerebral Cortex , Forkhead Transcription Factors , Hypoxia-Inducible Factor 1, alpha Subunit , Neural Stem Cells , Repressor Proteins , Signal Transduction , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Animals , Mice , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Repressor Proteins/metabolism , Repressor Proteins/genetics , Neovascularization, Physiologic/genetics , Cell Differentiation/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Neurogenesis/genetics , Glycolysis , Angiogenesis
6.
Nature ; 629(8010): 146-153, 2024 May.
Article En | MEDLINE | ID: mdl-38632406

Astrocytes, the most abundant non-neuronal cell type in the mammalian brain, are crucial circuit components that respond to and modulate neuronal activity through calcium (Ca2+) signalling1-7. Astrocyte Ca2+ activity is highly heterogeneous and occurs across multiple spatiotemporal scales-from fast, subcellular activity3,4 to slow, synchronized activity across connected astrocyte networks8-10-to influence many processes5,7,11. However, the inputs that drive astrocyte network dynamics remain unclear. Here we used ex vivo and in vivo two-photon astrocyte imaging while mimicking neuronal neurotransmitter inputs at multiple spatiotemporal scales. We find that brief, subcellular inputs of GABA and glutamate lead to widespread, long-lasting astrocyte Ca2+ responses beyond an individual stimulated cell. Further, we find that a key subset of Ca2+ activity-propagative activity-differentiates astrocyte network responses to these two main neurotransmitters, and may influence responses to future inputs. Together, our results demonstrate that local, transient neurotransmitter inputs are encoded by broad cortical astrocyte networks over a minutes-long time course, contributing to accumulating evidence that substantial astrocyte-neuron communication occurs across slow, network-level spatiotemporal scales12-14. These findings will enable future studies to investigate the link between specific astrocyte Ca2+ activity and specific functional outputs, which could build a consistent framework for astrocytic modulation of neuronal activity.


Astrocytes , Cerebral Cortex , Glutamic Acid , Nerve Net , Neurotransmitter Agents , gamma-Aminobutyric Acid , Animals , Female , Male , Mice , Astrocytes/metabolism , Astrocytes/cytology , Calcium/metabolism , Calcium Signaling , Cell Communication , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , gamma-Aminobutyric Acid/metabolism , Glutamic Acid/metabolism , Mice, Inbred C57BL , Nerve Net/cytology , Nerve Net/metabolism , Neurons/metabolism , Neurotransmitter Agents/metabolism , Time Factors
7.
J Neurosci Methods ; 406: 110137, 2024 Jun.
Article En | MEDLINE | ID: mdl-38626853

BACKGROUND: The neuronal and gliaI populations within the brain are tightly interwoven, making isolation and study of large populations of a single cell type from brain tissue a major technical challenge. Concurrently, cell-type specific extracellular vesicles (EVs) hold enormous diagnostic and therapeutic potential in neurodegenerative disorders including Alzheimer's disease (AD). NEW METHOD: Postmortem AD cortical samples were thawed and gently dissociated. Following filtration, myelin and red blood cell removal, cell pellets were immunolabeled with fluorescent antibodies and analyzed by flow cytometry. The cell pellet supernatant was applied to a triple sucrose cushion for brain EV isolation. RESULTS: Neuronal, astrocyte and microglial cell populations were identified. Cell integrity was demonstrated using calcein AM, which is retained by cells with esterase activity and an intact membrane. For some experiments cell pellets were fixed, permeabilized, and immunolabeled for cell-specific markers. Characterization of brain small EV fractions showed the expected size, depletion of EV negative markers, and enrichment in positive and cell-type specific markers. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS: We optimized and integrated established protocols, aiming to maximize information obtained from each human autopsy brain sample. The uniqueness of our method lies in its capability to isolate cells and EVs from a single cryopreserved brain sample. Our results not only demonstrate the feasibility of isolating specific brain cell subpopulations for RNA-seq but also validate these subpopulations at the protein level. The accelerated study of EVs from human samples is crucial for a better understanding of their contribution to neuron/glial crosstalk and disease progression.


Alzheimer Disease , Cerebral Cortex , Cryopreservation , Extracellular Vesicles , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Extracellular Vesicles/metabolism , Cryopreservation/methods , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Neurons/metabolism , Aged , Male , Female , Astrocytes/metabolism , Aged, 80 and over , Cell Separation/methods , Flow Cytometry/methods , Microglia/metabolism
8.
Sci Rep ; 14(1): 9846, 2024 04 29.
Article En | MEDLINE | ID: mdl-38684715

Astrocytes are glycolytically active cells in the central nervous system playing a crucial role in various brain processes from homeostasis to neurotransmission. Astrocytes possess a complex branched morphology, frequently examined by fluorescent microscopy. However, staining and fixation may impact the properties of astrocytes, thereby affecting the accuracy of the experimental data of astrocytes dynamics and morphology. On the other hand, phase contrast microscopy can be used to study astrocytes morphology without affecting them, but the post-processing of the resulting low-contrast images is challenging. The main result of this work is a novel approach for recognition and morphological analysis of unstained astrocytes based on machine-learning recognition of microscopic images. We conducted a series of experiments involving the cultivation of isolated astrocytes from the rat brain cortex followed by microscopy. Using the proposed approach, we tracked the temporal evolution of the average total length of branches, branching, and area per astrocyte in our experiments. We believe that the proposed approach and the obtained experimental data will be of interest and benefit to the scientific communities in cell biology, biophysics, and machine learning.


Astrocytes , Machine Learning , Microscopy, Phase-Contrast , Astrocytes/cytology , Animals , Microscopy, Phase-Contrast/methods , Rats , Cells, Cultured , Image Processing, Computer-Assisted/methods , Cerebral Cortex/cytology
9.
Neuroreport ; 35(8): 542-550, 2024 May 15.
Article En | MEDLINE | ID: mdl-38597273

Wnt signaling plays an important role in adult brain function, and its dysregulation has been implicated in the loss of neuronal homeostasis. Despite the existence of many studies on the participation of the Wnt pathway in adult neurons, its regulation in astrocytes has been scarcely explored. Several reports point to the presence of Wnt ligands in astrocytes and their possible impact on neuronal plasticity or neuronal death. We aimed to analyze the effect of the neurotransmitter glutamate and the inflammatory cytokine TNFα on the mRNA and protein levels of the canonical Wnt agonist Wnt7a and the antagonist Dkk1 in cultured astrocytes. Primary astrocyte cultures from rat cerebral cortices were exposed to glutamate or TNFα. Wnt7a and Dkk1 expression was analyzed by RT-qPCR and its protein abundance and distribution was assessed by immunofluorescence. We found high basal expression and protein levels of Wnt7a and Dkk1 in unstimulated astrocytes and overproduction of Dkk1 mRNA induced by the two stimuli. These results reveal the astrocytic source of the canonical Wnt ligands Wnt7a and Dkk1, whose levels are differentially regulated by glutamate and TNFα. Astrocytes are a significant source of Wnt ligands, the production of which can be differentially regulated under excitatory or proinflammatory conditions, thereby impacting neuronal function.


Astrocytes , Glutamic Acid , Intercellular Signaling Peptides and Proteins , Proto-Oncogene Proteins , Tumor Necrosis Factor-alpha , Wnt Proteins , Astrocytes/metabolism , Astrocytes/drug effects , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Glutamic Acid/metabolism , Wnt Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Rats , RNA, Messenger/metabolism , Rats, Wistar , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/cytology
10.
Brain Stimul ; 17(2): 365-381, 2024.
Article En | MEDLINE | ID: mdl-38492885

BACKGROUND: Intracortical microstimulation (ICMS) is used to map neuronal circuitry in the brain and restore lost sensory function, including vision, hearing, and somatosensation. The temporal response of cortical neurons to single pulse ICMS is remarkably stereotyped and comprises short latency excitation followed by prolonged inhibition and, in some cases, rebound excitation. However, the neural origin of the different response components to ICMS are poorly understood, and the interactions between the three response components during trains of ICMS pulses remains unclear. OBJECTIVE: We used computational modeling to determine the mechanisms contributing to the temporal response to ICMS in model cortical neurons. METHODS: We implemented a biophysically based computational model of a cortical column comprising neurons with realistic morphology and synapses and quantified the temporal response of cortical neurons to different ICMS protocols. We characterized the temporal responses to single pulse ICMS across stimulation intensities and inhibitory (GABA-B/GABA-A) synaptic strengths. To probe interactions between response components, we quantified the response to paired pulse ICMS at different inter-pulse intervals and the response to short trains at different stimulation frequencies. Finally, we evaluated the performance of biomimetic ICMS trains in evoking sustained neural responses. RESULTS: Single pulse ICMS evoked short latency excitation followed by a period of inhibition, but model neurons did not exhibit post-inhibitory excitation. The strength of short latency excitation increased and the duration of inhibition increased with increased stimulation amplitude. Prolonged inhibition resulted from both after-hyperpolarization currents and GABA-B synaptic transmission. During the paired pulse protocol, the strength of short latency excitation evoked by a test pulse decreased marginally compared to those evoked by a single pulse for interpulse intervals (IPI) < 100 m s. Further, the duration of inhibition evoked by the test pulse was prolonged compared to single pulse for IPIs <50 m s and was not predicted by linear superposition of individual inhibitory responses. For IPIs>50 m s, the duration of inhibition evoked by the test pulse was comparable to those evoked by a single pulse. Short ICMS trains evoked repetitive excitatory responses against a background of inhibition. However, the strength of the repetitive excitatory response declined during ICMS at higher frequencies. Further, the duration of inhibition at the cessation of ICMS at higher frequencies was prolonged compared to the duration following a single pulse. Biomimetic pulse trains evoked comparable neural response between the onset and offset phases despite the presence of stimulation induced inhibition. CONCLUSIONS: The cortical column model replicated the short latency excitation and long-lasting inhibitory components of the stereotyped neural response documented in experimental studies of ICMS. Both cellular and synaptic mechanisms influenced the response components generated by ICMS. The non-linear interactions between response components resulted in dynamic ICMS-evoked neural activity and may play an important role in mediating the ICMS-induced precepts.


Cerebral Cortex , Electric Stimulation , Models, Neurological , Neurons , Neurons/physiology , Electric Stimulation/methods , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Animals , Computer Simulation
11.
Nat Methods ; 21(5): 908-913, 2024 May.
Article En | MEDLINE | ID: mdl-38514779

Mapping neuronal networks from three-dimensional electron microscopy (3D-EM) data still poses substantial reconstruction challenges, in particular for thin axons. Currently available automated image segmentation methods require manual proofreading for many types of connectomic analysis. Here we introduce RoboEM, an artificial intelligence-based self-steering 3D 'flight' system trained to navigate along neurites using only 3D-EM data as input. Applied to 3D-EM data from mouse and human cortex, RoboEM substantially improves automated state-of-the-art segmentations and can replace manual proofreading for more complex connectomic analysis problems, yielding computational annotation cost for cortical connectomes about 400-fold lower than the cost of manual error correction.


Connectome , Imaging, Three-Dimensional , Synapses , Connectome/methods , Animals , Mice , Humans , Imaging, Three-Dimensional/methods , Synapses/physiology , Synapses/ultrastructure , Microscopy, Electron/methods , Artificial Intelligence , Algorithms , Image Processing, Computer-Assisted/methods , Cerebral Cortex/cytology
12.
Glia ; 72(7): 1290-1303, 2024 Jul.
Article En | MEDLINE | ID: mdl-38506330

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.


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
13.
Neuron ; 112(9): 1426-1443.e11, 2024 May 01.
Article En | MEDLINE | ID: mdl-38442714

Glucocorticoids are important for proper organ maturation, and their levels are tightly regulated during development. Here, we use human cerebral organoids and mice to study the cell-type-specific effects of glucocorticoids on neurogenesis. We show that glucocorticoids increase a specific type of basal progenitors (co-expressing PAX6 and EOMES) that has been shown to contribute to cortical expansion in gyrified species. This effect is mediated via the transcription factor ZBTB16 and leads to increased production of neurons. A phenome-wide Mendelian randomization analysis of an enhancer variant that moderates glucocorticoid-induced ZBTB16 levels reveals causal relationships with higher educational attainment and altered brain structure. The relationship with postnatal cognition is also supported by data from a prospective pregnancy cohort study. This work provides a cellular and molecular pathway for the effects of glucocorticoids on human neurogenesis that relates to lasting postnatal phenotypes.


Cerebral Cortex , Glucocorticoids , Neurogenesis , Promyelocytic Leukemia Zinc Finger Protein , Neurogenesis/drug effects , Neurogenesis/physiology , Humans , Animals , Mice , Glucocorticoids/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Female , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Pregnancy , Neurons/metabolism , Neurons/drug effects , Organoids/drug effects , Organoids/metabolism , Gene Expression Regulation, Developmental/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Male
14.
Pflugers Arch ; 476(5): 721-733, 2024 May.
Article En | MEDLINE | ID: mdl-38376567

Since more than a century, neuroscientists have distinguished excitatory (glutamatergic) neurons with long-distance projections from inhibitory (GABAergic) neurons with local projections and established layer-dependent schemes for the ~ 80% excitatory (principal) cells as well as the ~ 20% inhibitory neurons. Whereas, in the early days, mainly morphological criteria were used to define cell types, later supplemented by electrophysiological and neurochemical properties, nowadays. single-cell transcriptomics is the method of choice for cell type classification. Bringing recent insight together, we conclude that despite all established layer- and area-dependent differences, there is a set of reliably identifiable cortical cell types that were named (among others) intratelencephalic (IT), extratelencephalic (ET), and corticothalamic (CT) for the excitatory cells, which altogether comprise ~ 56 transcriptomic cell types (t-types). By the same means, inhibitory neurons were subdivided into parvalbumin (PV), somatostatin (SST), vasoactive intestinal polypeptide (VIP), and "other (i.e. Lamp5/Sncg)" subpopulations, which altogether comprise ~ 60 t-types. The coming years will show which t-types actually translate into "real" cell types that show a common set of multimodal features, including not only transcriptome but also physiology and morphology as well as connectivity and ultimately function. Only with the better knowledge of clear-cut cell types and experimental access to them, we will be able to reveal their specific functions, a task which turned out to be difficult in a part of the brain being so much specialized for cognition as the cerebral cortex.


Cerebral Cortex , Neurons , Animals , Neurons/metabolism , Neurons/physiology , Neurons/classification , Humans , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Cerebral Cortex/cytology , Transcriptome
15.
Nature ; 627(8002): 149-156, 2024 Mar.
Article En | MEDLINE | ID: mdl-38418876

The glymphatic movement of fluid through the brain removes metabolic waste1-4. Noninvasive 40 Hz stimulation promotes 40 Hz neural activity in multiple brain regions and attenuates pathology in mouse models of Alzheimer's disease5-8. Here we show that multisensory gamma stimulation promotes the influx of cerebrospinal fluid and the efflux of interstitial fluid in the cortex of the 5XFAD mouse model of Alzheimer's disease. Influx of cerebrospinal fluid was associated with increased aquaporin-4 polarization along astrocytic endfeet and dilated meningeal lymphatic vessels. Inhibiting glymphatic clearance abolished the removal of amyloid by multisensory 40 Hz stimulation. Using chemogenetic manipulation and a genetically encoded sensor for neuropeptide signalling, we found that vasoactive intestinal peptide interneurons facilitate glymphatic clearance by regulating arterial pulsatility. Our findings establish novel mechanisms that recruit the glymphatic system to remove brain amyloid.


Alzheimer Disease , Amyloid , Brain , Cerebrospinal Fluid , Extracellular Fluid , Gamma Rhythm , Glymphatic System , Animals , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid/metabolism , Aquaporin 4/metabolism , Astrocytes/metabolism , Brain/cytology , Brain/metabolism , Brain/pathology , Cerebrospinal Fluid/metabolism , Disease Models, Animal , Extracellular Fluid/metabolism , Glymphatic System/physiology , Interneurons/metabolism , Vasoactive Intestinal Peptide/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Electric Stimulation
16.
J Biol Chem ; 300(2): 105629, 2024 Feb.
Article En | MEDLINE | ID: mdl-38199563

In contrast to stage-specific transcription factors, the role of ubiquitous transcription factors in neuronal development remains a matter of scrutiny. Here, we demonstrated that a ubiquitous factor NF-Y is essential for neural progenitor maintenance during brain morphogenesis. Deletion of the NF-YA subunit in neural progenitors by using nestin-cre transgene in mice resulted in significant abnormalities in brain morphology, including a thinner cerebral cortex and loss of striatum during embryogenesis. Detailed analyses revealed a progressive decline in multiple neural progenitors in the cerebral cortex and ganglionic eminences, accompanied by induced apoptotic cell death and reduced cell proliferation. In neural progenitors, the NF-YA short isoform lacking exon 3 is dominant and co-expressed with cell cycle genes. ChIP-seq analysis from the cortex during early corticogenesis revealed preferential binding of NF-Y to the cell cycle genes, some of which were confirmed to be downregulated following NF-YA deletion. Notably, the NF-YA short isoform disappears and is replaced by its long isoform during neuronal differentiation. Forced expression of the NF-YA long isoform in neural progenitors resulted in a significant decline in neuronal count, possibly due to the suppression of cell proliferation. Collectively, we elucidated a critical role of the NF-YA short isoform in maintaining neural progenitors, possibly by regulating cell proliferation and apoptosis. Moreover, we identified an isoform switch in NF-YA within the neuronal lineage in vivo, which may explain the stage-specific role of NF-Y during neuronal development.


CCAAT-Binding Factor , Cerebral Cortex , Animals , Mice , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Gene Expression Regulation , Neurogenesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/metabolism
17.
Eur J Pharmacol ; 964: 176306, 2024 Feb 05.
Article En | MEDLINE | ID: mdl-38145647

During the production of orexin A and B from preproorexin, a common precursor protein, in hypothalamic orexin neurons, C-terminal peptide (herein called preproorexin C-peptide) is concomitantly produced via post-translational processing. The predicted three-dimensional structure of preproorexin C-peptide is similar among mammalian species, suggestive of a conserved function in the mammalian brain. However, C-peptide has long been regarded as a non-functional peptide. We herein examined the effects of rat and/or mouse preproorexin C-peptide on gene expression and cell viability in cultured rat cerebrocortical cells and on memory behavior in C57BL/6J mice. Rat and mouse C-peptides both increased brain-derived neurotrophic factor (Bdnf) mRNA levels. Moreover, C-peptide enhanced high K+-, glutamate-, and BDNF-induced increases in Bdnf mRNA levels without affecting forskolin-induced Bdnf expression. H-89, a protein kinase A inhibitor, blocked C-peptide-induced Bdnf expression, whereas rolipram, a phosphodiesterase inhibitor, enhanced this effect. Intracellular cyclic AMP concentrations were elevated by C-peptide. These results demonstrate that preproorexin C-peptide promoted Bdnf mRNA expression by a cyclic AMP-dependent mechanism. Eleven amino acids at the N terminus of rat preproorexin C-peptide exerted similar effects on Bdnf expression as full-length preproorexin C-peptide. Preproorexin C-peptide also exerted protective effects against CoCl2-induced neuronal cell death. An intracerebroventricular injection of mouse preproorexin C-peptide induced c-fos and Bdnf expression in the cerebral cortex and hippocampus and enhanced novel object recognition memory in mice. Collectively, the present results show that preproorexin C-peptide is a functional substance, at least in some pharmacological and neuronal settings.


Brain-Derived Neurotrophic Factor , Cerebral Cortex , Orexins , Peptide Fragments , Animals , Mice , Rats , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP/metabolism , Mice, Inbred C57BL , Orexins/pharmacology , RNA, Messenger/metabolism , Peptide Fragments/pharmacology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism
18.
Nature ; 624(7991): 403-414, 2023 Dec.
Article En | MEDLINE | ID: mdl-38092914

The brain controls nearly all bodily functions via spinal projecting neurons (SPNs) that carry command signals from the brain to the spinal cord. However, a comprehensive molecular characterization of brain-wide SPNs is still lacking. Here we transcriptionally profiled a total of 65,002 SPNs, identified 76 region-specific SPN types, and mapped these types into a companion atlas of the whole mouse brain1. This taxonomy reveals a three-component organization of SPNs: (1) molecularly homogeneous excitatory SPNs from the cortex, red nucleus and cerebellum with somatotopic spinal terminations suitable for point-to-point communication; (2) heterogeneous populations in the reticular formation with broad spinal termination patterns, suitable for relaying commands related to the activities of the entire spinal cord; and (3) modulatory neurons expressing slow-acting neurotransmitters and/or neuropeptides in the hypothalamus, midbrain and reticular formation for 'gain setting' of brain-spinal signals. In addition, this atlas revealed a LIM homeobox transcription factor code that parcellates the reticulospinal neurons into five molecularly distinct and spatially segregated populations. Finally, we found transcriptional signatures of a subset of SPNs with large soma size and correlated these with fast-firing electrophysiological properties. Together, this study establishes a comprehensive taxonomy of brain-wide SPNs and provides insight into the functional organization of SPNs in mediating brain control of bodily functions.


Brain , Gene Expression Profiling , Neural Pathways , Neurons , Spinal Cord , Animals , Mice , Hypothalamus , Neurons/metabolism , Neuropeptides , Spinal Cord/cytology , Spinal Cord/metabolism , Brain/cytology , Brain/metabolism , Neurotransmitter Agents , Mesencephalon/cytology , Reticular Formation/cytology , Electrophysiology , Cerebellum/cytology , Cerebral Cortex/cytology
19.
Nature ; 624(7991): 378-389, 2023 Dec.
Article En | MEDLINE | ID: mdl-38092917

Recent advances in single-cell technologies have led to the discovery of thousands of brain cell types; however, our understanding of the gene regulatory programs in these cell types is far from complete1-4. Here we report a comprehensive atlas of candidate cis-regulatory DNA elements (cCREs) in the adult mouse brain, generated by analysing chromatin accessibility in 2.3 million individual brain cells from 117 anatomical dissections. The atlas includes approximately 1 million cCREs and their chromatin accessibility across 1,482 distinct brain cell populations, adding over 446,000 cCREs to the most recent such annotation in the mouse genome. The mouse brain cCREs are moderately conserved in the human brain. The mouse-specific cCREs-specifically, those identified from a subset of cortical excitatory neurons-are strongly enriched for transposable elements, suggesting a potential role for transposable elements in the emergence of new regulatory programs and neuronal diversity. Finally, we infer the gene regulatory networks in over 260 subclasses of mouse brain cells and develop deep-learning models to predict the activities of gene regulatory elements in different brain cell types from the DNA sequence alone. Our results provide a resource for the analysis of cell-type-specific gene regulation programs in both mouse and human brains.


Brain , Chromatin , Single-Cell Analysis , Animals , Humans , Mice , Brain/cytology , Brain/metabolism , Cerebral Cortex/cytology , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Deep Learning , DNA Transposable Elements/genetics , Gene Regulatory Networks/genetics , Neurons/metabolism
20.
Nature ; 622(7981): 120-129, 2023 Oct.
Article En | MEDLINE | ID: mdl-37674083

Multimodal astrocyte-neuron communications govern brain circuitry assembly and function1. For example, through rapid glutamate release, astrocytes can control excitability, plasticity and synchronous activity2,3 of synaptic networks, while also contributing to their dysregulation in neuropsychiatric conditions4-7. For astrocytes to communicate through fast focal glutamate release, they should possess an apparatus for Ca2+-dependent exocytosis similar to neurons8-10. However, the existence of this mechanism has been questioned11-13 owing to inconsistent data14-17 and a lack of direct supporting evidence. Here we revisited the astrocyte glutamate exocytosis hypothesis by considering the emerging molecular heterogeneity of astrocytes18-21 and using molecular, bioinformatic and imaging approaches, together with cell-specific genetic tools that interfere with glutamate exocytosis in vivo. By analysing existing single-cell RNA-sequencing databases and our patch-seq data, we identified nine molecularly distinct clusters of hippocampal astrocytes, among which we found a notable subpopulation that selectively expressed synaptic-like glutamate-release machinery and localized to discrete hippocampal sites. Using GluSnFR-based glutamate imaging22 in situ and in vivo, we identified a corresponding astrocyte subgroup that responds reliably to astrocyte-selective stimulations with subsecond glutamate release events at spatially precise hotspots, which were suppressed by astrocyte-targeted deletion of vesicular glutamate transporter 1 (VGLUT1). Furthermore, deletion of this transporter or its isoform VGLUT2 revealed specific contributions of glutamatergic astrocytes in cortico-hippocampal and nigrostriatal circuits during normal behaviour and pathological processes. By uncovering this atypical subpopulation of specialized astrocytes in the adult brain, we provide insights into the complex roles of astrocytes in central nervous system (CNS) physiology and diseases, and identify a potential therapeutic target.


Astrocytes , Central Nervous System , Glutamic Acid , Signal Transduction , Adult , Humans , Astrocytes/classification , Astrocytes/cytology , Astrocytes/metabolism , Central Nervous System/cytology , Central Nervous System/metabolism , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Neurons/metabolism , Synaptic Transmission , Calcium/metabolism , Exocytosis , Single-Cell Gene Expression Analysis , Vesicular Glutamate Transport Protein 1/deficiency , Vesicular Glutamate Transport Protein 1/genetics , Gene Deletion , Cerebral Cortex/cytology , Cerebral Cortex/metabolism
...