Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22.903
1.
Methods Mol Biol ; 2799: 139-150, 2024.
Article En | MEDLINE | ID: mdl-38727906

Epilepsy is one of the most represented neurological diseases worldwide. However, in many cases, the precise molecular mechanisms of epileptogenesis and ictiogenesis are unknown. Because of their important role in synaptic function and neuronal excitability, NMDA receptors are implicated in various epileptogenic mechanisms. Most of these are subunit specific and require a precise analysis of the subunit composition of the NMDARs implicated. Here, we describe an express electrophysiological method to analyze the contribution of NMDAR subunits to spontaneous postsynaptic activity in identified cells in brain slices using patch clamp whole cell recordings.


Patch-Clamp Techniques , Receptors, N-Methyl-D-Aspartate , Synapses , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Patch-Clamp Techniques/methods , Synapses/metabolism , Synapses/physiology , Brain/metabolism , Brain/cytology , Neurons/metabolism , Mice , Rats , Protein Subunits/metabolism
2.
Brief Bioinform ; 25(3)2024 Mar 27.
Article En | MEDLINE | ID: mdl-38739758

The complicated process of neuronal development is initiated early in life, with the genetic mechanisms governing this process yet to be fully elucidated. Single-cell RNA sequencing (scRNA-seq) is a potent instrument for pinpointing biomarkers that exhibit differential expression across various cell types and developmental stages. By employing scRNA-seq on human embryonic stem cells, we aim to identify differentially expressed genes (DEGs) crucial for early-stage neuronal development. Our focus extends beyond simply identifying DEGs. We strive to investigate the functional roles of these genes through enrichment analysis and construct gene regulatory networks to understand their interactions. Ultimately, this comprehensive approach aspires to illuminate the molecular mechanisms and transcriptional dynamics governing early human brain development. By uncovering potential links between these DEGs and intelligence, mental disorders, and neurodevelopmental disorders, we hope to shed light on human neurological health and disease. In this study, we have used scRNA-seq to identify DEGs involved in early-stage neuronal development in hESCs. The scRNA-seq data, collected on days 26 (D26) and 54 (D54), of the in vitro differentiation of hESCs to neurons were analyzed. Our analysis identified 539 DEGs between D26 and D54. Functional enrichment of those DEG biomarkers indicated that the up-regulated DEGs participated in neurogenesis, while the down-regulated DEGs were linked to synapse regulation. The Reactome pathway analysis revealed that down-regulated DEGs were involved in the interactions between proteins located in synapse pathways. We also discovered interactions between DEGs and miRNA, transcriptional factors (TFs) and DEGs, and between TF and miRNA. Our study identified 20 significant transcription factors, shedding light on early brain development genetics. The identified DEGs and gene regulatory networks are valuable resources for future research into human brain development and neurodevelopmental disorders.


Biomarkers , Brain , Gene Regulatory Networks , Human Embryonic Stem Cells , Single-Cell Analysis , Humans , Single-Cell Analysis/methods , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Brain/metabolism , Brain/embryology , Brain/cytology , Biomarkers/metabolism , Neurons/metabolism , Neurons/cytology , Cell Differentiation/genetics , RNA-Seq , Neurogenesis/genetics , Gene Expression Regulation, Developmental , Gene Expression Profiling , Sequence Analysis, RNA/methods , Single-Cell Gene Expression Analysis
3.
Methods Mol Biol ; 2800: 203-215, 2024.
Article En | MEDLINE | ID: mdl-38709486

Cell tracking is an essential step in extracting cellular signals from moving cells, which is vital for understanding the mechanisms underlying various biological functions and processes, particularly in organs such as the brain and heart. However, cells in living organisms often exhibit extensive and complex movements caused by organ deformation and whole-body motion. These movements pose a challenge in obtaining high-quality time-lapse cell images and tracking the intricate cell movements in the captured images. Recent advances in deep learning techniques provide powerful tools for detecting cells in low-quality images with densely packed cell populations, as well as estimating cell positions for cells undergoing large nonrigid movements. This chapter introduces the challenges of cell tracking in deforming organs and moving animals, outlines the solutions to these challenges, and presents a detailed protocol for data preparation, as well as for performing cell segmentation and tracking using the latest version of 3DeeCellTracker. This protocol is expected to enable researchers to gain deeper insights into organ dynamics and biological processes.


Cell Tracking , Deep Learning , Animals , Cell Tracking/methods , Image Processing, Computer-Assisted/methods , Cell Movement , Brain/cytology , Time-Lapse Imaging/methods
4.
Sci Data ; 11(1): 514, 2024 May 20.
Article En | MEDLINE | ID: mdl-38769371

Brain organoids represent a useful tool for modeling of neurodevelopmental disorders and can recapitulate brain volume alterations such as microcephaly. To monitor organoid growth, brightfield microscopy images are frequently used and evaluated manually which is time-consuming and prone to observer-bias. Recent software applications for organoid evaluation address this issue using classical or AI-based methods. These pipelines have distinct strengths and weaknesses that are not evident to external observers. We provide a dataset of more than 1,400 images of 64 trackable brain organoids from four clones differentiated from healthy and diseased patients. This dataset is especially powerful to test and compare organoid analysis pipelines because of (1) trackable organoids (2) frequent imaging during development (3) clone diversity (4) distinct clone development (5) cross sample imaging by two different labs (6) common imaging distractors, and (6) pixel-level ground truth organoid annotations. Therefore, this dataset allows to perform differentiated analyses to delineate strengths, weaknesses, and generalizability of automated organoid analysis pipelines as well as analysis of clone diversity and similarity.


Brain , Organoids , Organoids/cytology , Brain/diagnostic imaging , Brain/cytology , Humans
5.
Mol Brain ; 17(1): 24, 2024 May 18.
Article En | MEDLINE | ID: mdl-38762724

CD11c-positive (CD11c+) microglia have attracted considerable attention because of their potential implications in central nervous system (CNS) development, homeostasis, and disease. However, the spatiotemporal dynamics of the proportion of CD11c+ microglia in individual CNS regions are poorly understood. Here, we investigated the proportion of CD11c+ microglia in six CNS regions (forebrain, olfactory bulb, diencephalon/midbrain, cerebellum, pons/medulla, and spinal cord) from the developmental to adult stages by flow cytometry and immunohistochemical analyses using a CD11c reporter transgenic mouse line, Itgax-Venus. We found that the proportion of CD11c+ microglia in total microglia varied between CNS regions during postnatal development. Specifically, the proportion was high in the olfactory bulb and cerebellum at postnatal day P(4) and P7, respectively, and approximately half of the total microglia were CD11c+. The proportion declined sharply in all regions to P14, and the low percentage persisted over P56. In the spinal cord, the proportion of CD11c+ microglia was also high at P4 and declined to P14, but increased again at P21 and thereafter. Interestingly, the distribution pattern of CD11c+ microglia in the spinal cord markedly changed from gray matter at P4 to white matter at P21. Collectively, our findings reveal the differences in the spatiotemporal dynamics of the proportion of CD11c+ microglia among CNS regions from early development to adult stages in normal mice. These findings improve our understanding of the nature of microglial heterogeneity and its dynamics in the CNS.


Brain , Mice, Transgenic , Microglia , Spinal Cord , Animals , Microglia/metabolism , Microglia/cytology , Spinal Cord/growth & development , Brain/growth & development , Brain/cytology , Spatio-Temporal Analysis , Aging , CD11c Antigen/metabolism , Mice, Inbred C57BL , Mice , Animals, Newborn
6.
Nat Commun ; 15(1): 4102, 2024 May 22.
Article En | MEDLINE | ID: mdl-38778027

The development of robust tools for segmenting cellular and sub-cellular neuronal structures lags behind the massive production of high-resolution 3D images of neurons in brain tissue. The challenges are principally related to high neuronal density and low signal-to-noise characteristics in thick samples, as well as the heterogeneity of data acquired with different imaging methods. To address this issue, we design a framework which includes sample preparation for high resolution imaging and image analysis. Specifically, we set up a method for labeling thick samples and develop SENPAI, a scalable algorithm for segmenting neurons at cellular and sub-cellular scales in conventional and super-resolution STimulated Emission Depletion (STED) microscopy images of brain tissues. Further, we propose a validation paradigm for testing segmentation performance when a manual ground-truth may not exhaustively describe neuronal arborization. We show that SENPAI provides accurate multi-scale segmentation, from entire neurons down to spines, outperforming state-of-the-art tools. The framework will empower image processing of complex neuronal circuitries.


Algorithms , Brain , Imaging, Three-Dimensional , Neurons , Neurons/cytology , Animals , Brain/diagnostic imaging , Brain/cytology , Imaging, Three-Dimensional/methods , Mice , Image Processing, Computer-Assisted/methods
7.
Cell Rep Methods ; 4(5): 100777, 2024 May 20.
Article En | MEDLINE | ID: mdl-38744289

Human brain tissue models and organoids are vital for studying and modeling human neurological disease. However, the high cost of long-term cultured organoids inhibits their wide-ranging application. It is therefore urgent to develop methods for the cryopreservation of brain tissue and organoids. Here, we establish a method using methylcellulose, ethylene glycol, DMSO, and Y27632 (termed MEDY) for the cryopreservation of cortical organoids without disrupting the neural cytoarchitecture or functional activity. MEDY can be applied to multiple brain-region-specific organoids, including the dorsal/ventral forebrain, spinal cord, optic vesicle brain, and epilepsy patient-derived brain organoids. Additionally, MEDY enables the cryopreservation of human brain tissue samples, and pathological features are retained after thawing. Transcriptomic analysis shows that MEDY can protect synaptic function and inhibit the endoplasmic reticulum-mediated apoptosis pathway. MEDY will enable the large-scale and reliable storage of diverse neural organoids and living brain tissue and will facilitate wide-ranging research, medical applications, and drug screening.


Brain , Cryopreservation , Organoids , Humans , Organoids/drug effects , Cryopreservation/methods , Brain/drug effects , Brain/cytology , Neurons/drug effects , Neurons/physiology , Ethylene Glycol/pharmacology , Methylcellulose/chemistry , Methylcellulose/pharmacology , Dimethyl Sulfoxide/pharmacology
8.
Nat Cell Biol ; 26(5): 710-718, 2024 May.
Article En | MEDLINE | ID: mdl-38714853

During brain development, neural progenitors expand through symmetric divisions before giving rise to differentiating cell types via asymmetric divisions. Transition between those modes varies among individual neural stem cells, resulting in clones of different sizes. Imaging-based lineage tracing allows for lineage analysis at high cellular resolution but systematic approaches to analyse clonal behaviour of entire tissues are currently lacking. Here we implement whole-tissue lineage tracing by genomic DNA barcoding in 3D human cerebral organoids, to show that individual stem cell clones produce progeny on a vastly variable scale. By using stochastic modelling we find that variable lineage sizes arise because a subpopulation of lineages retains symmetrically dividing cells. We show that lineage sizes can adjust to tissue demands after growth perturbation via chemical ablation or genetic restriction of a subset of cells in chimeric organoids. Our data suggest that adaptive plasticity of stem cell populations ensures robustness of development in human brain organoids.


Cell Lineage , Neural Stem Cells , Organoids , Organoids/cytology , Organoids/metabolism , Humans , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Differentiation , Cell Proliferation , Clone Cells , Neurogenesis/genetics , DNA Barcoding, Taxonomic , Animals
9.
Trends Neurosci ; 47(5): 322-323, 2024 May.
Article En | MEDLINE | ID: mdl-38664110

In a recent study, Shvedov and colleagues used live two-photon imaging in transgenic zebra finches to reveal migration patterns of neuroblasts through the complex environment of the postembryonic brain. This study highlights the value of ubiquitin C/green fluorescent protein (UBC-GFP) transgenic zebra finches in studying adult neurogenesis and advances our understanding of dispersed long-distance neuronal migration in the adult brain, shedding light on this understudied phenomenon.


Brain , Cell Movement , Neurogenesis , Neurons , Songbirds , Animals , Cell Movement/physiology , Neurogenesis/physiology , Brain/physiology , Brain/cytology , Neurons/physiology , Songbirds/physiology , Animals, Genetically Modified , Finches/physiology , Neural Stem Cells/physiology
10.
PLoS Comput Biol ; 20(4): e1012054, 2024 Apr.
Article En | MEDLINE | ID: mdl-38648250

Neural organoids model the development of the human brain and are an indispensable tool for studying neurodevelopment. Whole-organoid lineage tracing has revealed the number of progenies arising from each initial stem cell to be highly diverse, with lineage sizes ranging from one to more than 20,000 cells. This high variability exceeds what can be explained by existing stochastic models of corticogenesis and indicates the existence of an additional source of stochasticity. To explain this variability, we introduce the SAN model which distinguishes Symmetrically diving, Asymmetrically dividing, and Non-proliferating cells. In the SAN model, the additional source of stochasticity is the survival time of a lineage's pool of symmetrically dividing cells. These survival times result from neutral competition within the sub-population of all symmetrically dividing cells. We demonstrate that our model explains the experimentally observed variability of lineage sizes and derive the quantitative relationship between survival time and lineage size. We also show that our model implies the existence of a regulatory mechanism which keeps the size of the symmetrically dividing cell population constant. Our results provide quantitative insight into the clonal composition of neural organoids and how it arises. This is relevant for many applications of neural organoids, and similar processes may occur in other developing tissues both in vitro and in vivo.


Organoids , Organoids/cytology , Humans , Cell Lineage/physiology , Computational Biology , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Stochastic Processes , Models, Biological , Neurons/physiology , Neurons/cytology , Brain/cytology , Brain/physiology , Cell Proliferation/physiology , Neurogenesis/physiology
11.
Nat Rev Neurosci ; 25(6): 414-427, 2024 Jun.
Article En | MEDLINE | ID: mdl-38658739

Microglia constitute the largest population of parenchymal macrophages in the brain and are considered a unique subset of central nervous system glial cells owing to their extra-embryonic origins in the yolk sac. During development, microglial progenitors readily proliferate and eventually colonize the entire brain. In this Review, we highlight the origins of microglial progenitors and their entry routes into the brain and discuss the various molecular and non-molecular determinants of their fate, which may inform their specific functions. Specifically, we explore recently identified mechanisms that regulate microglial colonization of the brain, including the availability of space, and describe how the expansion of highly proliferative microglial progenitors facilitates the occupation of the microglial niche. Finally, we shed light on the factors involved in establishing microglial identity in the brain.


Microglia , Microglia/physiology , Microglia/metabolism , Animals , Humans , Brain/growth & development , Brain/cytology , Brain/metabolism , Cell Differentiation/physiology
12.
Open Biol ; 14(4): 230383, 2024 Apr.
Article En | MEDLINE | ID: mdl-38629124

Non-clustered protocadherins (ncPcdhs) are adhesive molecules with spatio-temporally regulated overlapping expression in the developing nervous system. Although their unique role in neurogenesis has been widely studied, their combinatorial role in brain physiology and pathology is poorly understood. Using probabilistic cell typing by in situ sequencing, we demonstrate combinatorial inter- and intra-familial expression of ncPcdhs in the developing mouse cortex and hippocampus, at single-cell resolution. We discovered the combinatorial expression of Protocadherin-19 (Pcdh19), a protein involved in PCDH19-clustering epilepsy, with Pcdh1, Pcdh9 or Cadherin 13 (Cdh13) in excitatory neurons. Using aggregation assays, we demonstrate a code-specific adhesion function of PCDH19; mosaic PCDH19 absence in PCDH19+9 and PCDH19 + CDH13, but not in PCDH19+1 codes, alters cell-cell interaction. Interestingly, we found that PCDH19 as a dominant protein in two heterophilic adhesion codes could promote trans-interaction between them. In addition, we discovered increased CDH13-mediated cell adhesion in the presence of PCDH19, suggesting a potential role of PCDH19 as an adhesion mediator of CDH13. Finally, we demonstrated novel cis-interactions between PCDH19 and PCDH1, PCDH9 and CDH13. These observations suggest that there is a unique combinatorial code with a cell- and region-specific characteristic where a single molecule defines the heterophilic cell-cell adhesion properties of each code.


Brain , Cell Adhesion , Protocadherins , Animals , Mice , Brain/cytology , Brain/growth & development , Epilepsy/metabolism , Neurons/metabolism
13.
Nature ; 628(8009): 863-871, 2024 Apr.
Article En | MEDLINE | ID: mdl-38570687

Vertebrate organs require locally adapted blood vessels1,2. The gain of such organotypic vessel specializations is often deemed to be molecularly unrelated to the process of organ vascularization. Here, opposing this model, we reveal a molecular mechanism for brain-specific angiogenesis that operates under the control of Wnt7a/b ligands-well-known blood-brain barrier maturation signals3-5. The control mechanism relies on Wnt7a/b-dependent expression of Mmp25, which we find is enriched in brain endothelial cells. CRISPR-Cas9 mutagenesis in zebrafish reveals that this poorly characterized glycosylphosphatidylinositol-anchored matrix metalloproteinase is selectively required in endothelial tip cells to enable their initial migration across the pial basement membrane lining the brain surface. Mechanistically, Mmp25 confers brain invasive competence by cleaving meningeal fibroblast-derived collagen IV α5/6 chains within a short non-collagenous region of the central helical part of the heterotrimer. After genetic interference with the pial basement membrane composition, the Wnt-ß-catenin-dependent organotypic control of brain angiogenesis is lost, resulting in properly patterned, yet blood-brain-barrier-defective cerebrovasculatures. We reveal an organ-specific angiogenesis mechanism, shed light on tip cell mechanistic angiodiversity and thereby illustrate how organs, by imposing local constraints on angiogenic tip cells, can select vessels matching their distinctive physiological requirements.


Brain , Neovascularization, Physiologic , Animals , Basement Membrane/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/cytology , Brain/cytology , Brain/blood supply , Brain/metabolism , Cell Movement , Collagen Type IV/metabolism , CRISPR-Cas Systems/genetics , Endothelial Cells/metabolism , Endothelial Cells/cytology , Meninges/cytology , Meninges/blood supply , Meninges/metabolism , Organ Specificity , Wnt Proteins/metabolism , Wnt Signaling Pathway , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics
15.
Cell ; 187(9): 2143-2157.e15, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38670072

A central question for regenerative neuroscience is whether synthetic neural circuits, such as those built from two species, can function in an intact brain. Here, we apply blastocyst complementation to selectively build and test interspecies neural circuits. Despite approximately 10-20 million years of evolution, and prominent species differences in brain size, rat pluripotent stem cells injected into mouse blastocysts develop and persist throughout the mouse brain. Unexpectedly, the mouse niche reprograms the birth dates of rat neurons in the cortex and hippocampus, supporting rat-mouse synaptic activity. When mouse olfactory neurons are genetically silenced or killed, rat neurons restore information flow to odor processing circuits. Moreover, they rescue the primal behavior of food seeking, although less well than mouse neurons. By revealing that a mouse can sense the world using neurons from another species, we establish neural blastocyst complementation as a powerful tool to identify conserved mechanisms of brain development, plasticity, and repair.


Neurons , Animals , Mice , Rats , Neurons/metabolism , Neurons/cytology , Neurons/physiology , Blastocyst/metabolism , Blastocyst/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Brain/cytology , Brain/physiology , Female , Hippocampus/cytology , Hippocampus/physiology , Species Specificity , Mice, Inbred C57BL , Male
16.
Genome Biol ; 25(1): 109, 2024 Apr 26.
Article En | MEDLINE | ID: mdl-38671451

Single-cell multiplexing techniques (cell hashing and genetic multiplexing) combine multiple samples, optimizing sample processing and reducing costs. Cell hashing conjugates antibody-tags or chemical-oligonucleotides to cell membranes, while genetic multiplexing allows to mix genetically diverse samples and relies on aggregation of RNA reads at known genomic coordinates. We develop hadge (hashing deconvolution combined with genotype information), a Nextflow pipeline that combines 12 methods to perform both hashing- and genotype-based deconvolution. We propose a joint deconvolution strategy combining best-performing methods and demonstrate how this approach leads to the recovery of previously discarded cells in a nuclei hashing of fresh-frozen brain tissue.


Single-Cell Analysis , Single-Cell Analysis/methods , Humans , Brain/metabolism , Brain/cytology , Software , Genotype
17.
Cells ; 13(8)2024 Apr 12.
Article En | MEDLINE | ID: mdl-38667283

Astrocytes and ependymal cells have been reported to be able to switch from a mature cell identity towards that of a neural stem/progenitor cell. Astrocytes are widely scattered in the brain where they exert multiple functions and are routinely targeted for in vitro and in vivo reprogramming. Ependymal cells serve more specialized functions, lining the ventricles and the central canal, and are multiciliated, epithelial-like cells that, in the spinal cord, act as bi-potent progenitors in response to injury. Here, we isolate or generate ependymal cells and post-mitotic astrocytes, respectively, from the lateral ventricles of the mouse brain and we investigate their capacity to reverse towards a progenitor-like identity in culture. Inhibition of the GSK3 and TGFß pathways facilitates the switch of mature astrocytes to Sox2-expressing, mitotic cells that generate oligodendrocytes. Although this medium allows for the expansion of quiescent NSCs, isolated from live rats by "milking of the brain", it does not fully reverse astrocytes towards the bona fide NSC identity; this is a failure correlated with a concomitant lack of neurogenic activity. Ependymal cells could be induced to enter mitosis either via exposure to neuraminidase-dependent stress or by culturing them in the presence of FGF2 and EGF. Overall, our data confirm that astrocytes and ependymal cells retain a high capacity to reverse to a progenitor identity and set up a simple and highly controlled platform for the elucidation of the molecular mechanisms that regulate this reversal.


Astrocytes , Ependyma , Phenotype , Animals , Astrocytes/metabolism , Astrocytes/cytology , Ependyma/cytology , Ependyma/metabolism , Mice , Cells, Cultured , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Cell Differentiation , Brain/cytology , Brain/metabolism , Rats , SOXB1 Transcription Factors/metabolism , Mice, Inbred C57BL , Mitosis , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Animals, Newborn
18.
J Pharmacol Sci ; 155(2): 29-34, 2024 Jun.
Article En | MEDLINE | ID: mdl-38677783

Microglia are the residential immune cells in the central nervous system. Their roles as innate immune cells and regulators of synaptic remodeling are critical to the development and the maintenance of the brain. Numerous studies have depleted microglia to elucidate their involvement in healthy and pathological conditions. PLX3397, a blocker of colony stimulating factor 1 receptor (CSF1R), is widely used to deplete mouse microglia due to its non-invasiveness and convenience. Recently, other small rodents, including Syrian hamsters (Mesocricetus auratus) and Mongolian gerbils (Meriones unguiculatus), have been recognized as valuable animal models for studying brain functions and diseases. However, whether microglia depletion via PLX3397 is feasible in these species remains unclear. Here, we administered PLX3397 orally via food pellets to hamsters and gerbils. PLX3397 successfully depleted gerbil microglia but had no effect on microglial density in hamsters. Comparative analysis of the CSF1R amino acid sequence in different species hints that amino acid substitutions in the juxtamembrane domain may potentially contribute to the inefficacy of PLX3397 in hamsters.


Aminopyridines , Gerbillinae , Mesocricetus , Microglia , Pyrroles , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor , Animals , Microglia/drug effects , Microglia/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Pyrroles/pharmacology , Male , Cricetinae , Pyrrolidines/pharmacology , Species Specificity , Models, Animal , Administration, Oral , Brain/drug effects , Brain/metabolism , Brain/cytology
19.
Nat Neurosci ; 27(5): 846-861, 2024 May.
Article En | MEDLINE | ID: mdl-38539013

The generation of new myelin-forming oligodendrocytes in the adult central nervous system is critical for cognitive function and regeneration following injury. Oligodendrogenesis varies between gray and white matter regions, suggesting that local cues drive regional differences in myelination and the capacity for regeneration. However, the layer- and region-specific regulation of oligodendrocyte populations is unclear due to the inability to monitor deep brain structures in vivo. Here we harnessed the superior imaging depth of three-photon microscopy to permit long-term, longitudinal in vivo three-photon imaging of the entire cortical column and subcortical white matter in adult mice. We find that cortical oligodendrocyte populations expand at a higher rate in the adult brain than those of the white matter. Following demyelination, oligodendrocyte replacement is enhanced in the white matter, while the deep cortical layers show deficits in regenerative oligodendrogenesis and the restoration of transcriptional heterogeneity. Together, our findings demonstrate that regional microenvironments regulate oligodendrocyte population dynamics and heterogeneity in the healthy and diseased brain.


Oligodendroglia , White Matter , Animals , Oligodendroglia/physiology , Mice , White Matter/physiology , Demyelinating Diseases/pathology , Myelin Sheath/physiology , Mice, Inbred C57BL , Male , Mice, Transgenic , Nerve Regeneration/physiology , Female , Brain/physiology , Brain/cytology , Neurogenesis/physiology
20.
Nature ; 628(8007): 381-390, 2024 Apr.
Article En | MEDLINE | ID: mdl-38480888

Our understanding of the neurobiology of primate behaviour largely derives from artificial tasks in highly controlled laboratory settings, overlooking most natural behaviours that primate brains evolved to produce1-3. How primates navigate the multidimensional social relationships that structure daily life4 and shape survival and reproductive success5 remains largely unclear at the single-neuron level. Here we combine ethological analysis, computer vision and wireless recording technologies to identify neural signatures of natural behaviour in unrestrained, socially interacting pairs of rhesus macaques. Single-neuron and population activity in the prefrontal and temporal cortex robustly encoded 24 species-typical behaviours, as well as social context. Male-female partners demonstrated near-perfect reciprocity in grooming, a key behavioural mechanism supporting friendships and alliances6, and neural activity maintained a running account of these social investments. Confronted with an aggressive intruder, behavioural and neural population responses reflected empathy and were buffered by the presence of a partner. Our findings reveal a highly distributed neurophysiological ledger of social dynamics, a potential computational foundation supporting communal life in primate societies, including our own.


Brain , Macaca mulatta , Neurons , Social Behavior , Animals , Female , Male , Aggression/physiology , Brain/cytology , Brain/physiology , Empathy , Grooming , Group Processes , Macaca mulatta/classification , Macaca mulatta/physiology , Macaca mulatta/psychology , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Temporal Lobe/cytology , Temporal Lobe/physiology , Neurons/physiology
...