Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Nature ; 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693260

ABSTRACT

The human brain develops through a tightly organized cascade of patterning events, induced by transcription factor expression and changes in chromatin accessibility. Although gene expression across the developing brain has been described at single-cell resolution1, similar atlases of chromatin accessibility have been primarily focused on the forebrain2-4. Here we describe chromatin accessibility and paired gene expression across the entire developing human brain during the first trimester (6-13 weeks after conception). We defined 135 clusters and used multiomic measurements to link candidate cis-regulatory elements to gene expression. The number of accessible regions increased both with age and along neuronal differentiation. Using a convolutional neural network, we identified putative functional transcription factor-binding sites in enhancers characterizing neuronal subtypes. We applied this model to cis-regulatory elements linked to ESRRB to elucidate its activation mechanism in the Purkinje cell lineage. Finally, by linking disease-associated single nucleotide polymorphisms to cis-regulatory elements, we validated putative pathogenic mechanisms in several diseases and identified midbrain-derived GABAergic neurons as being the most vulnerable to major depressive disorder-related mutations. Our findings provide a more detailed view of key gene regulatory mechanisms underlying the emergence of brain cell types during the first trimester and a comprehensive reference for future studies related to human neurodevelopment.

2.
Science ; 382(6667): eadf1226, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824650

ABSTRACT

The adult human brain comprises more than a thousand distinct neuronal and glial cell types, a diversity that emerges during early brain development. To reveal the precise sequence of events during early brain development, we used single-cell RNA sequencing and spatial transcriptomics and uncovered cell states and trajectories in human brains at 5 to 14 postconceptional weeks (pcw). We identified 12 major classes that are organized as ~600 distinct cell states, which map to precise spatial anatomical domains at 5 pcw. We described detailed differentiation trajectories of the human forebrain and midbrain and found a large number of region-specific glioblasts that mature into distinct pre-astrocytes and pre-oligodendrocyte precursor cells. Our findings reveal the establishment of cell types during the first trimester of human brain development.


Subject(s)
Brain , Neurogenesis , Pregnancy Trimester, First , Female , Humans , Pregnancy , Astrocytes/cytology , Brain/cytology , Brain/embryology , Neuroglia , Neurons/cytology , Atlases as Topic , Single-Cell Gene Expression Analysis
3.
Science ; 382(6667): eadd7046, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824663

ABSTRACT

The human brain directs complex behaviors, ranging from fine motor skills to abstract intelligence, but the diversity of cell types that support these skills has not been fully described. In this work, we used single-nucleus RNA sequencing to systematically survey cells across the entire adult human brain. We sampled more than three million nuclei from approximately 100 dissections across the forebrain, midbrain, and hindbrain in three postmortem donors. Our analysis identified 461 clusters and 3313 subclusters organized largely according to developmental origins and revealing high diversity in midbrain and hindbrain neurons. Astrocytes and oligodendrocyte-lineage cells also exhibited regional diversity at multiple scales. The transcriptomic census of the entire human brain presented in this work provides a resource for understanding the molecular diversity of the human brain in health and disease.


Subject(s)
Brain , Transcriptome , Adult , Humans , Brain/cytology , Brain/metabolism , Gene Expression Profiling , Mesencephalon , Neurons/metabolism , Prosencephalon , Single-Cell Gene Expression Analysis
4.
Nature ; 596(7870): 92-96, 2021 08.
Article in English | MEDLINE | ID: mdl-34321664

ABSTRACT

The mammalian brain develops through a complex interplay of spatial cues generated by diffusible morphogens, cell-cell interactions and intrinsic genetic programs that result in probably more than a thousand distinct cell types. A complete understanding of this process requires a systematic characterization of cell states over the entire spatiotemporal range of brain development. The ability of single-cell RNA sequencing and spatial transcriptomics to reveal the molecular heterogeneity of complex tissues has therefore been particularly powerful in the nervous system. Previous studies have explored development in specific brain regions1-8, the whole adult brain9 and even entire embryos10. Here we report a comprehensive single-cell transcriptomic atlas of the embryonic mouse brain between gastrulation and birth. We identified almost eight hundred cellular states that describe a developmental program for the functional elements of the brain and its enclosing membranes, including the early neuroepithelium, region-specific secondary organizers, and both neurogenic and gliogenic progenitors. We also used in situ mRNA sequencing to map the spatial expression patterns of key developmental genes. Integrating the in situ data with our single-cell clusters revealed the precise spatial organization of neural progenitors during the patterning of the nervous system.


Subject(s)
Brain/cytology , Brain/embryology , Single-Cell Analysis , Transcriptome , Animals , Animals, Newborn/genetics , Brain/anatomy & histology , Female , Gastrulation/genetics , Male , Mice , Neural Tube/anatomy & histology , Neural Tube/cytology , Neural Tube/embryology
6.
Nat Med ; 27(4): 640-646, 2021 04.
Article in English | MEDLINE | ID: mdl-33859435

ABSTRACT

Apart from well-defined factors in neuronal cells1, only a few reports consider that the variability of sporadic amyotrophic lateral sclerosis (ALS) progression can depend on less-defined contributions from glia2,3 and blood vessels4. In this study we use an expression-weighted cell-type enrichment method to infer cell activity in spinal cord samples from patients with sporadic ALS and mouse models of this disease. Here we report that patients with sporadic ALS present cell activity patterns consistent with two mouse models in which enrichments of vascular cell genes preceded microglial response. Notably, during the presymptomatic stage, perivascular fibroblast cells showed the strongest gene enrichments, and their marker proteins SPP1 and COL6A1 accumulated in enlarged perivascular spaces in patients with sporadic ALS. Moreover, in plasma of 574 patients with ALS from four independent cohorts, increased levels of SPP1 at disease diagnosis repeatedly predicted shorter survival with stronger effect than the established risk factors of bulbar onset or neurofilament levels in cerebrospinal fluid. We propose that the activity of the recently discovered perivascular fibroblast can predict survival of patients with ALS and provide a new conceptual framework to re-evaluate definitions of ALS etiology.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , Blood Vessels/pathology , Fibroblasts/pathology , Amyotrophic Lateral Sclerosis/blood , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Biomarkers/metabolism , Collagen Type VI/genetics , Collagen Type VI/metabolism , DNA-Binding Proteins/metabolism , Disease Progression , Genetic Markers , Humans , Mice, Transgenic , Osteopontin/blood , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spinal Cord/pathology , Spinal Cord/ultrastructure , Superoxide Dismutase/genetics , Transcription, Genetic , Vascular Remodeling
7.
Nat Biotechnol ; 39(8): 968-977, 2021 08.
Article in English | MEDLINE | ID: mdl-33875865

ABSTRACT

Several techniques are currently being developed for spatially resolved omics profiling, but each new method requires the setup of specific detection strategies or specialized instrumentation. Here we describe an imaging-free framework to localize high-throughput readouts within a tissue by cutting the sample into thin strips in a way that allows subsequent image reconstruction. We implemented this framework to transform a low-input RNA sequencing protocol into an imaging-free spatial transcriptomics technique (called STRP-seq) and validated it by profiling the spatial transcriptome of the mouse brain. We applied the technique to the brain of the Australian bearded dragon, Pogona vitticeps. Our results reveal the molecular anatomy of the telencephalon of this lizard, providing evidence for a marked regionalization of the reptilian pallium and subpallium. We expect that STRP-seq can be used to derive spatially resolved data from a range of other omics techniques.


Subject(s)
Gene Expression Profiling/methods , Molecular Imaging/methods , Tomography/methods , Algorithms , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Chemistry , Lizards , Mice , Transcriptome/genetics
8.
Nat Commun ; 12(1): 1510, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33686078

ABSTRACT

Distinct types of dorsal root ganglion sensory neurons may have unique contributions to chronic pain. Identification of primate sensory neuron types is critical for understanding the cellular origin and heritability of chronic pain. However, molecular insights into the primate sensory neurons are missing. Here we classify non-human primate dorsal root ganglion sensory neurons based on their transcriptome and map human pain heritability to neuronal types. First, we identified cell correlates between two major datasets for mouse sensory neuron types. Machine learning exposes an overall cross-species conservation of somatosensory neurons between primate and mouse, although with differences at individual gene level, highlighting the importance of primate data for clinical translation. We map genomic loci associated with chronic pain in human onto primate sensory neuron types to identify the cellular origin of chronic pain. Genome-wide associations for chronic pain converge on two different neuronal types distributed between pain disorders that display different genetic susceptibilities, suggesting both unique and shared mechanisms between different pain conditions.


Subject(s)
Chronic Pain/genetics , Chronic Pain/metabolism , Sensory Receptor Cells/metabolism , Transcriptome , Animals , Female , Ganglia, Spinal , Gene Expression , Humans , Macaca mulatta , Male , Mice , Neurons , Primates
9.
Cell Rep ; 31(5): 107601, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32375051

ABSTRACT

Liver X receptors (LXRs) and their ligands are potent regulators of midbrain dopaminergic (mDA) neurogenesis and differentiation. However, the molecular mechanisms by which LXRs control these functions remain to be elucidated. Here, we perform a combined transcriptome and chromatin immunoprecipitation sequencing (ChIP-seq) analysis of midbrain cells after LXR activation, followed by bioinformatic analysis to elucidate the transcriptional networks controlling mDA neurogenesis. Our results identify the basic helix-loop-helix transcription factor sterol regulatory element binding protein 1 (SREBP1) as part of a cluster of proneural transcription factors in radial glia and as a regulator of transcription factors controlling mDA neurogenesis, such as Foxa2. Moreover, loss- and gain-of-function experiments in vitro and in vivo demonstrate that Srebf1 is both required and sufficient for mDA neurogenesis. Our data, thus, identify Srebf1 as a central player in mDA neurogenesis.


Subject(s)
Cell Differentiation/physiology , Dopaminergic Neurons/metabolism , Neurogenesis/physiology , Sterol Regulatory Element Binding Protein 1/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Dopamine/metabolism , Gene Expression Regulation, Developmental/physiology , Humans , Mesencephalon/cytology , Nerve Tissue Proteins/metabolism
10.
Nature ; 560(7719): 494-498, 2018 08.
Article in English | MEDLINE | ID: mdl-30089906

ABSTRACT

RNA abundance is a powerful indicator of the state of individual cells. Single-cell RNA sequencing can reveal RNA abundance with high quantitative accuracy, sensitivity and throughput1. However, this approach captures only a static snapshot at a point in time, posing a challenge for the analysis of time-resolved phenomena such as embryogenesis or tissue regeneration. Here we show that RNA velocity-the time derivative of the gene expression state-can be directly estimated by distinguishing between unspliced and spliced mRNAs in common single-cell RNA sequencing protocols. RNA velocity is a high-dimensional vector that predicts the future state of individual cells on a timescale of hours. We validate its accuracy in the neural crest lineage, demonstrate its use on multiple published datasets and technical platforms, reveal the branching lineage tree of the developing mouse hippocampus, and examine the kinetics of transcription in human embryonic brain. We expect RNA velocity to greatly aid the analysis of developmental lineages and cellular dynamics, particularly in humans.


Subject(s)
Brain/cytology , Neural Crest/metabolism , Neurons/cytology , RNA Splicing/genetics , RNA/analysis , RNA/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Animals , Brain/embryology , Brain/metabolism , Cell Lineage/genetics , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Datasets as Topic , Female , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/metabolism , Kinetics , Male , Mice , Neural Crest/cytology , Neurons/metabolism , Reproducibility of Results , Time Factors , Transcription, Genetic/genetics
11.
Cell Rep ; 24(8): 2179-2190.e7, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30134177

ABSTRACT

Striatal locally projecting neurons, or interneurons, act on nearby circuits and shape functional output to the rest of the basal ganglia. We performed single-cell RNA sequencing of striatal cells enriching for interneurons. We find seven discrete interneuron types, six of which are GABAergic. In addition to providing specific markers for the populations previously described, including those expressing Sst/Npy, Th, Npy without Sst, and Chat, we identify two small populations of cells expressing Cck with or without Vip. Surprisingly, the Pvalb-expressing cells do not constitute a discrete cluster but rather are part of a larger group of cells expressing Pthlh with a spatial gradient of Pvalb expression. Using PatchSeq, we show that Pthlh cells exhibit a continuum of electrophysiological properties correlated with expression of Pvalb. Furthermore, we find significant molecular differences that correlate with differences in electrophysiological properties between Pvalb-expressing cells of the striatum and those of the cortex.


Subject(s)
Corpus Striatum/metabolism , Interneurons/metabolism , Sequence Analysis, RNA/methods , Animals , Humans , Mice
12.
Development ; 145(18)2018 09 25.
Article in English | MEDLINE | ID: mdl-30126905

ABSTRACT

Touch and mechanical sensations require the development of several different kinds of sensory neurons dedicated to respond to certain types of mechanical stimuli. The transcription factor Shox2 (short stature homeobox 2) is involved in the generation of TRKB+ low-threshold mechanoreceptors (LTMRs), but mechanisms terminating this program and allowing alternative fates are unknown. Here, we show that the conditional loss of the miR-183-96-182 cluster in mouse leads to a failure of extinction of Shox2 during development and an increase in the proportion of Aδ LTMRs (TRKB+/NECAB2+) neurons at the expense of Aß slowly adapting (SA)-LTMRs (TRKC+/Runx3-) neurons. Conversely, overexpression of miR-183 cluster that represses Shox2 expression, or loss of Shox2, both increase the Aß SA-LTMRs population at the expense of Aδ LTMRs. Our results suggest that the miR-183 cluster determines the timing of Shox2 expression by direct targeting during development, and through this determines the population sizes of Aδ LTMRs and Aß SA-LTMRs.


Subject(s)
Homeodomain Proteins/metabolism , Mechanoreceptors/metabolism , MicroRNAs/genetics , Sensory Receptor Cells/cytology , Animals , Calcium-Binding Proteins/metabolism , Cell Differentiation/genetics , Eye Proteins/metabolism , Female , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Pregnancy , Protein-Tyrosine Kinases/metabolism
13.
Cell ; 174(4): 999-1014.e22, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30096314

ABSTRACT

The mammalian nervous system executes complex behaviors controlled by specialized, precisely positioned, and interacting cell types. Here, we used RNA sequencing of half a million single cells to create a detailed census of cell types in the mouse nervous system. We mapped cell types spatially and derived a hierarchical, data-driven taxonomy. Neurons were the most diverse and were grouped by developmental anatomical units and by the expression of neurotransmitters and neuropeptides. Neuronal diversity was driven by genes encoding cell identity, synaptic connectivity, neurotransmission, and membrane conductance. We discovered seven distinct, regionally restricted astrocyte types that obeyed developmental boundaries and correlated with the spatial distribution of key glutamate and glycine neurotransmitters. In contrast, oligodendrocytes showed a loss of regional identity followed by a secondary diversification. The resource presented here lays a solid foundation for understanding the molecular architecture of the mammalian nervous system and enables genetic manipulation of specific cell types.


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Nervous System/metabolism , Single-Cell Analysis/methods , Transcriptome , Animals , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Male , Mice , Mice, Inbred C57BL , Nervous System/growth & development
14.
Nat Neurosci ; 21(6): 869-880, 2018 06.
Article in English | MEDLINE | ID: mdl-29686262

ABSTRACT

The dorsal horn of the spinal cord is critical to processing distinct modalities of noxious and innocuous sensation, but little is known of the neuronal subtypes involved, hampering efforts to deduce principles governing somatic sensation. Here we used single-cell RNA sequencing to classify sensory neurons in the mouse dorsal horn. We identified 15 inhibitory and 15 excitatory molecular subtypes of neurons, equaling the complexity in cerebral cortex. Validating our classification scheme in vivo and matching cell types to anatomy of the dorsal horn by spatial transcriptomics reveals laminar enrichment for each of the cell types. Neuron types, when combined, define a multilayered organization with like neurons layered together. Employing our scheme, we find that heat and cold stimuli activate discrete sets of both excitatory and inhibitory neuron types. This work provides a systematic and comprehensive molecular classification of spinal cord sensory neurons, enabling functional interrogation of sensory processing.


Subject(s)
Atlases as Topic , Neurons/physiology , Sensation/physiology , Spinal Cord Dorsal Horn/physiology , Transcriptome/genetics , Animals , Cold Temperature , Female , Glutamates/physiology , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Neurons/classification , Posterior Horn Cells/physiology , RNA/genetics , Sensory Receptor Cells/classification , Sensory Receptor Cells/physiology , Spinal Cord/cytology , Spinal Cord/physiology , Spinal Cord Dorsal Horn/anatomy & histology
15.
Nat Neurosci ; 21(2): 290-299, 2018 02.
Article in English | MEDLINE | ID: mdl-29335606

ABSTRACT

The dentate gyrus of the hippocampus is a brain region in which neurogenesis persists into adulthood; however, the relationship between developmental and adult dentate gyrus neurogenesis has not been examined in detail. Here we used single-cell RNA sequencing to reveal the molecular dynamics and diversity of dentate gyrus cell types in perinatal, juvenile, and adult mice. We found distinct quiescent and proliferating progenitor cell types, linked by transient intermediate states to neuroblast stages and fully mature granule cells. We observed shifts in the molecular identity of quiescent and proliferating radial glia and granule cells during the postnatal period that were then maintained through adult stages. In contrast, intermediate progenitor cells, neuroblasts, and immature granule cells were nearly indistinguishable at all ages. These findings demonstrate the fundamental similarity of postnatal and adult neurogenesis in the hippocampus and pinpoint the early postnatal transformation of radial glia from embryonic progenitors to adult quiescent stem cells.


Subject(s)
Base Sequence/physiology , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Neurogenesis/physiology , Neurons/physiology , Animals , Animals, Newborn , Cell Differentiation , Cell Proliferation , Chromium/metabolism , Female , Fructose-Bisphosphate Aldolase/metabolism , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Neuroglia/physiology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Single-Cell Analysis
16.
Sci Rep ; 7(1): 16327, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180631

ABSTRACT

Single-cell RNA-seq has become routine for discovering cell types and revealing cellular diversity, but archived human brain samples still pose a challenge to current high-throughput platforms. We present STRT-seq-2i, an addressable 9600-microwell array platform, combining sampling by limiting dilution or FACS, with imaging and high throughput at competitive cost. We applied the platform to fresh single mouse cortical cells and to frozen post-mortem human cortical nuclei, matching the performance of a previous lower-throughput platform while retaining a high degree of flexibility, potentially also for other high-throughput applications.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA , Single-Cell Analysis/methods , Animals , Computational Biology , Humans , Mice , RNA/genetics , RNA/isolation & purification , Sequence Analysis, RNA/methods , Workflow
17.
Mol Syst Biol ; 13(5): 930, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28495919

ABSTRACT

Alternative transcription start sites (TSSs) have been extensively studied genome-wide for many cell types and have been shown to be important during development and to regulate transcript abundance between cell types. Likewise, single-cell gene expression has been extensively studied for many cell types. However, how single cells use TSSs has not yet been examined. In particular, it is unknown whether alternative TSSs are independently expressed, or whether they are co-activated or even mutually exclusive in single cells. Here, we use a previously published single-cell RNA-seq dataset, comprising thousands of cells, to study alternative TSS usage. We find that alternative TSS usage is a regulated process, and the correlation between two TSSs expressed in single cells of the same cell type is surprisingly high. Our findings indicate that TSSs generally are regulated by common factors rather than being independently regulated or stochastically expressed.


Subject(s)
Brain/cytology , Transcription Initiation Site , Animals , Base Sequence , Cystatin C/genetics , Gene Expression , Mice , Single-Cell Analysis
18.
Cell ; 167(2): 566-580.e19, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27716510

ABSTRACT

Understanding human embryonic ventral midbrain is of major interest for Parkinson's disease. However, the cell types, their gene expression dynamics, and their relationship to commonly used rodent models remain to be defined. We performed single-cell RNA sequencing to examine ventral midbrain development in human and mouse. We found 25 molecularly defined human cell types, including five subtypes of radial glia-like cells and four progenitors. In the mouse, two mature fetal dopaminergic neuron subtypes diversified into five adult classes during postnatal development. Cell types and gene expression were generally conserved across species, but with clear differences in cell proliferation, developmental timing, and dopaminergic neuron development. Additionally, we developed a method to quantitatively assess the fidelity of dopaminergic neurons derived from human pluripotent stem cells, at a single-cell level. Thus, our study provides insight into the molecular programs controlling human midbrain development and provides a foundation for the development of cell replacement therapies.


Subject(s)
Dopaminergic Neurons/cytology , Mesencephalon/cytology , Mesencephalon/embryology , Neural Stem Cells/cytology , Neurogenesis , Pluripotent Stem Cells/cytology , Animals , Cell Line , Cellular Reprogramming Techniques , Humans , Machine Learning , Mesencephalon/metabolism , Mice , Neuroglia/cytology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
19.
Cell Syst ; 3(3): 221-237.e9, 2016 09 28.
Article in English | MEDLINE | ID: mdl-27641957

ABSTRACT

The murine epidermis with its hair follicles represents an invaluable model system for tissue regeneration and stem cell research. Here we used single-cell RNA-sequencing to reveal how cellular heterogeneity of murine telogen epidermis is tuned at the transcriptional level. Unbiased clustering of 1,422 single-cell transcriptomes revealed 25 distinct populations of interfollicular and follicular epidermal cells. Our data allowed the reconstruction of gene expression programs during epidermal differentiation and along the proximal-distal axis of the hair follicle at unprecedented resolution. Moreover, transcriptional heterogeneity of the epidermis can essentially be explained along these two axes, and we show that heterogeneity in stem cell compartments generally reflects this model: stem cell populations are segregated by spatial signatures but share a common basal-epidermal gene module. This study provides an unbiased and systematic view of transcriptional organization of adult epidermis and highlights how cellular heterogeneity can be orchestrated in vivo to assure tissue homeostasis.


Subject(s)
Transcriptome , Animals , Cell Differentiation , Epidermis , Female , Hair Follicle , Homeostasis , Mice , Mice, Inbred C57BL , Stem Cells
20.
Science ; 352(6291): 1326-1329, 2016 Jun 10.
Article in English | MEDLINE | ID: mdl-27284195

ABSTRACT

Oligodendrocytes have been considered as a functionally homogeneous population in the central nervous system (CNS). We performed single-cell RNA sequencing on 5072 cells of the oligodendrocyte lineage from 10 regions of the mouse juvenile and adult CNS. Thirteen distinct populations were identified, 12 of which represent a continuum from Pdgfra(+) oligodendrocyte precursor cells (OPCs) to distinct mature oligodendrocytes. Initial stages of differentiation were similar across the juvenile CNS, whereas subsets of mature oligodendrocytes were enriched in specific regions in the adult brain. Newly formed oligodendrocytes were detected in the adult CNS and were responsive to complex motor learning. A second Pdgfra(+) population, distinct from OPCs, was found along vessels. Our study reveals the dynamics of oligodendrocyte differentiation and maturation, uncoupling them at a transcriptional level and highlighting oligodendrocyte heterogeneity in the CNS.


Subject(s)
Brain/growth & development , Neurogenesis , Oligodendroglia/cytology , Animals , Antigens/genetics , Antigens/metabolism , Biomarkers/metabolism , Brain/cytology , Cell Lineage , Cells, Cultured , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Learning/physiology , Mice , Motor Activity/physiology , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , RNA, Messenger/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sequence Analysis, RNA , Single-Cell Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...