Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Science ; 384(6698): eadi5199, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781369

ABSTRACT

Single-cell genomics is a powerful tool for studying heterogeneous tissues such as the brain. Yet little is understood about how genetic variants influence cell-level gene expression. Addressing this, we uniformly processed single-nuclei, multiomics datasets into a resource comprising >2.8 million nuclei from the prefrontal cortex across 388 individuals. For 28 cell types, we assessed population-level variation in expression and chromatin across gene families and drug targets. We identified >550,000 cell type-specific regulatory elements and >1.4 million single-cell expression quantitative trait loci, which we used to build cell-type regulatory and cell-to-cell communication networks. These networks manifest cellular changes in aging and neuropsychiatric disorders. We further constructed an integrative model accurately imputing single-cell expression and simulating perturbations; the model prioritized ~250 disease-risk genes and drug targets with associated cell types.


Subject(s)
Brain , Gene Regulatory Networks , Mental Disorders , Single-Cell Analysis , Humans , Aging/genetics , Brain/metabolism , Cell Communication/genetics , Chromatin/metabolism , Chromatin/genetics , Genomics , Mental Disorders/genetics , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Quantitative Trait Loci
2.
bioRxiv ; 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38562822

ABSTRACT

Single-cell genomics is a powerful tool for studying heterogeneous tissues such as the brain. Yet, little is understood about how genetic variants influence cell-level gene expression. Addressing this, we uniformly processed single-nuclei, multi-omics datasets into a resource comprising >2.8M nuclei from the prefrontal cortex across 388 individuals. For 28 cell types, we assessed population-level variation in expression and chromatin across gene families and drug targets. We identified >550K cell-type-specific regulatory elements and >1.4M single-cell expression-quantitative-trait loci, which we used to build cell-type regulatory and cell-to-cell communication networks. These networks manifest cellular changes in aging and neuropsychiatric disorders. We further constructed an integrative model accurately imputing single-cell expression and simulating perturbations; the model prioritized ~250 disease-risk genes and drug targets with associated cell types.

3.
Science ; 382(6667): eade9516, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824638

ABSTRACT

The cognitive abilities of humans are distinctive among primates, but their molecular and cellular substrates are poorly understood. We used comparative single-nucleus transcriptomics to analyze samples of the middle temporal gyrus (MTG) from adult humans, chimpanzees, gorillas, rhesus macaques, and common marmosets to understand human-specific features of the neocortex. Human, chimpanzee, and gorilla MTG showed highly similar cell-type composition and laminar organization as well as a large shift in proportions of deep-layer intratelencephalic-projecting neurons compared with macaque and marmoset MTG. Microglia, astrocytes, and oligodendrocytes had more-divergent expression across species compared with neurons or oligodendrocyte precursor cells, and neuronal expression diverged more rapidly on the human lineage. Only a few hundred genes showed human-specific patterning, suggesting that relatively few cellular and molecular changes distinctively define adult human cortical structure.


Subject(s)
Cognition , Hominidae , Neocortex , Temporal Lobe , Animals , Humans , Gene Expression Profiling , Gorilla gorilla/genetics , Hominidae/genetics , Hominidae/physiology , Macaca mulatta/genetics , Pan troglodytes/genetics , Phylogeny , Transcriptome , Neocortex/physiology , Species Specificity , Temporal Lobe/physiology
4.
Science ; 382(6667): eadf2359, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824649

ABSTRACT

Single-cell transcriptomic studies have identified a conserved set of neocortical cell types from small postmortem cohorts. We extended these efforts by assessing cell type variation across 75 adult individuals undergoing epilepsy and tumor surgeries. Nearly all nuclei map to one of 125 robust cell types identified in the middle temporal gyrus. However, we found interindividual variance in abundances and gene expression signatures, particularly in deep-layer glutamatergic neurons and microglia. A minority of donor variance is explainable by age, sex, ancestry, disease state, and cell state. Genomic variation was associated with expression of 150 to 250 genes for most cell types. This characterization of cellular variation provides a baseline for cell typing in health and disease.


Subject(s)
Temporal Lobe , Transcriptome , Adult , Humans , Epilepsy/metabolism , Gene Expression Profiling , Neurons/metabolism , Temporal Lobe/cytology , Temporal Lobe/metabolism , Nervous System Diseases/genetics , Mental Disorders/genetics
5.
Science ; 382(6667): eadf6812, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824655

ABSTRACT

Variation in cytoarchitecture is the basis for the histological definition of cortical areas. We used single cell transcriptomics and performed cellular characterization of the human cortex to better understand cortical areal specialization. Single-nucleus RNA-sequencing of 8 areas spanning cortical structural variation showed a highly consistent cellular makeup for 24 cell subclasses. However, proportions of excitatory neuron subclasses varied substantially, likely reflecting differences in connectivity across primary sensorimotor and association cortices. Laminar organization of astrocytes and oligodendrocytes also differed across areas. Primary visual cortex showed characteristic organization with major changes in the excitatory to inhibitory neuron ratio, expansion of layer 4 excitatory neurons, and specialized inhibitory neurons. These results lay the groundwork for a refined cellular and molecular characterization of human cortical cytoarchitecture and areal specialization.


Subject(s)
Neocortex , Humans , Neocortex/metabolism , Neocortex/ultrastructure , Neurons/classification , Neurons/metabolism , Transcriptome , Single-Cell Gene Expression Analysis , Phylogeny
6.
Elife ; 122023 05 30.
Article in English | MEDLINE | ID: mdl-37249212

ABSTRACT

Rodent studies have demonstrated that synaptic dynamics from excitatory to inhibitory neuron types are often dependent on the target cell type. However, these target cell-specific properties have not been well investigated in human cortex, where there are major technical challenges in reliably obtaining healthy tissue, conducting multiple patch-clamp recordings on inhibitory cell types, and identifying those cell types. Here, we take advantage of newly developed methods for human neurosurgical tissue analysis with multiple patch-clamp recordings, post-hoc fluorescent in situ hybridization (FISH), machine learning-based cell type classification and prospective GABAergic AAV-based labeling to investigate synaptic properties between pyramidal neurons and PVALB- vs. SST-positive interneurons. We find that there are robust molecular differences in synapse-associated genes between these neuron types, and that individual presynaptic pyramidal neurons evoke postsynaptic responses with heterogeneous synaptic dynamics in different postsynaptic cell types. Using molecular identification with FISH and classifiers based on transcriptomically identified PVALB neurons analyzed by Patch-seq, we find that PVALB neurons typically show depressing synaptic characteristics, whereas other interneuron types including SST-positive neurons show facilitating characteristics. Together, these data support the existence of target cell-specific synaptic properties in human cortex that are similar to rodent, thereby indicating evolutionary conservation of local circuit connectivity motifs from excitatory to inhibitory neurons and their synaptic dynamics.


Subject(s)
Neocortex , Humans , Neocortex/physiology , Synaptic Transmission/physiology , In Situ Hybridization, Fluorescence , Prospective Studies , Neurons/physiology , Pyramidal Cells/physiology , Synapses/physiology , Interneurons/physiology
8.
Sci Data ; 10(1): 50, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36693887

ABSTRACT

Large-scale single-cell 'omics profiling is being used to define a complete catalogue of brain cell types, something that traditional methods struggle with due to the diversity and complexity of the brain. But this poses a problem: How do we organise such a catalogue - providing a standard way to refer to the cell types discovered, linking their classification and properties to supporting data? Cell ontologies provide a partial solution to these problems, but no existing ontology schemas support the definition of cell types by direct reference to supporting data, classification of cell types using classifications derived directly from data, or links from cell types to marker sets along with confidence scores. Here we describe a generally applicable schema that solves these problems and its application in a semi-automated pipeline to build a data-linked extension to the Cell Ontology representing cell types in the Primary Motor Cortex of humans, mice and marmosets. The methods and resulting ontology are designed to be scalable and applicable to similar whole-brain atlases currently in preparation.


Subject(s)
Biological Ontologies , Brain , Animals , Humans , Mice , Callithrix , Data Collection/standards
9.
Proc Natl Acad Sci U S A ; 119(48): e2202580119, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36417438

ABSTRACT

Neurons in the developing brain undergo extensive structural refinement as nascent circuits adopt their mature form. This physical transformation of neurons is facilitated by the engulfment and degradation of axonal branches and synapses by surrounding glial cells, including microglia and astrocytes. However, the small size of phagocytic organelles and the complex, highly ramified morphology of glia have made it difficult to define the contribution of these and other glial cell types to this crucial process. Here, we used large-scale, serial section transmission electron microscopy (TEM) with computational volume segmentation to reconstruct the complete 3D morphologies of distinct glial types in the mouse visual cortex, providing unprecedented resolution of their morphology and composition. Unexpectedly, we discovered that the fine processes of oligodendrocyte precursor cells (OPCs), a population of abundant, highly dynamic glial progenitors, frequently surrounded small branches of axons. Numerous phagosomes and phagolysosomes (PLs) containing fragments of axons and vesicular structures were present inside their processes, suggesting that OPCs engage in axon pruning. Single-nucleus RNA sequencing from the developing mouse cortex revealed that OPCs express key phagocytic genes at this stage, as well as neuronal transcripts, consistent with active axon engulfment. Although microglia are thought to be responsible for the majority of synaptic pruning and structural refinement, PLs were ten times more abundant in OPCs than in microglia at this stage, and these structures were markedly less abundant in newly generated oligodendrocytes, suggesting that OPCs contribute substantially to the refinement of neuronal circuits during cortical development.


Subject(s)
Neocortex , Oligodendrocyte Precursor Cells , Animals , Mice , Axons/metabolism , Oligodendroglia/metabolism , Neurons/metabolism
11.
Nature ; 598(7879): 111-119, 2021 10.
Article in English | MEDLINE | ID: mdl-34616062

ABSTRACT

The primary motor cortex (M1) is essential for voluntary fine-motor control and is functionally conserved across mammals1. Here, using high-throughput transcriptomic and epigenomic profiling of more than 450,000 single nuclei in humans, marmoset monkeys and mice, we demonstrate a broadly conserved cellular makeup of this region, with similarities that mirror evolutionary distance and are consistent between the transcriptome and epigenome. The core conserved molecular identities of neuronal and non-neuronal cell types allow us to generate a cross-species consensus classification of cell types, and to infer conserved properties of cell types across species. Despite the overall conservation, however, many species-dependent specializations are apparent, including differences in cell-type proportions, gene expression, DNA methylation and chromatin state. Few cell-type marker genes are conserved across species, revealing a short list of candidate genes and regulatory mechanisms that are responsible for conserved features of homologous cell types, such as the GABAergic chandelier cells. This consensus transcriptomic classification allows us to use patch-seq (a combination of whole-cell patch-clamp recordings, RNA sequencing and morphological characterization) to identify corticospinal Betz cells from layer 5 in non-human primates and humans, and to characterize their highly specialized physiology and anatomy. These findings highlight the robust molecular underpinnings of cell-type diversity in M1 across mammals, and point to the genes and regulatory pathways responsible for the functional identity of cell types and their species-specific adaptations.


Subject(s)
Motor Cortex/cytology , Neurons/classification , Single-Cell Analysis , Animals , Atlases as Topic , Callithrix/genetics , Epigenesis, Genetic , Epigenomics , Female , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Expression Profiling , Glutamates/metabolism , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Middle Aged , Motor Cortex/anatomy & histology , Neurons/cytology , Neurons/metabolism , Organ Specificity , Phylogeny , Species Specificity , Transcriptome
12.
Neuron ; 109(18): 2914-2927.e5, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34534454

ABSTRACT

In the neocortex, subcerebral axonal projections originate largely from layer 5 (L5) extratelencephalic-projecting (ET) neurons. The unique morpho-electric properties of these neurons have been mainly described in rodents, where retrograde tracers or transgenic lines can label them. Similar labeling strategies are infeasible in the human neocortex, rendering the translational relevance of findings in rodents unclear. We leveraged the recent discovery of a transcriptomically defined L5 ET neuron type to study the properties of human L5 ET neurons in neocortical brain slices derived from neurosurgeries. Patch-seq recordings, where transcriptome, physiology, and morphology were assayed from the same cell, revealed many conserved morpho-electric properties of human and rodent L5 ET neurons. Divergent properties were often subtler than differences between L5 cell types within these two species. These data suggest a conserved function of L5 ET neurons in the neocortical hierarchy but also highlight phenotypic divergence possibly related to functional specialization of human neocortex.


Subject(s)
Dendrites/physiology , Morphogenesis/physiology , Neocortex/cytology , Neocortex/physiology , Pyramidal Cells/physiology , Transcriptome/physiology , Action Potentials/physiology , Adult , Animals , Female , Humans , Macaca nemestrina , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Organ Culture Techniques , Patch-Clamp Techniques/methods
13.
Cell Rep ; 30(7): 2195-2208.e5, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32075759

ABSTRACT

Müller glia (MG) serve as sources for retinal regeneration in non-mammalian vertebrates. We find that this process can be induced in mouse MG, after injury, by transgenic expression of the proneural transcription factor Ascl1 and the HDAC inhibitor TSA. However, new neurons are generated only from a subset of MG. Identifying factors that limit Ascl1-mediated MG reprogramming could make this process more efficient. In this study, we test whether injury-induced STAT activation hampers the ability of Ascl1 to reprogram MG into retinal neurons. Single-cell RNA-seq shows that progenitor-like cells derived from Ascl1-expressing MG have a higher level of STAT signaling than do those cells that become neurons. Ascl1-ChIPseq and ATAC-seq show that STAT potentially directs Ascl1 to developmentally inappropriate targets. Using a STAT inhibitor, in combination with our previously described reprogramming paradigm, we found a large increase in the ability of MG to generate neurons.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin/metabolism , Nerve Regeneration/physiology , Neuroglia/physiology , Neurons/metabolism , STAT Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL , Neurogenesis , Signal Transduction
14.
Ann Med Surg (Lond) ; 41: 20-28, 2019 May.
Article in English | MEDLINE | ID: mdl-31011420

ABSTRACT

BACKGROUND: Optimizing nerve regeneration and mitigating muscle atrophy are the keys to successful outcomes in peripheral nerve damage. We investigated whether mesenchymal stem cell (MSC) therapy can improve limb function recovery in peripheral nerve damage. MATERIALS AND METHODS: We used sciatic nerve transection/repair (SNR) and individual nerve transection/repair (INR; branches of sciatic nerve - tibial, peroneal, sural) models to study the effect of MSCs on proximal and distal peripheral nerve damages, respectively, in male Lewis rats. Syngeneic MSCs (5 × 106; passage≤6) or saline were administered locally and intravenously. Sensory/motor functions (SF/MF) of the limb were assessed. RESULTS: Rat MSCs (>90%) were CD29+, CD90+, CD34-, CD31- and multipotent. Total SF at two weeks post-SNR & INR with or without MSC therapy was ∼1.2 on a 0-3 grading scale (0 = No function; 3 = Normal); by 12 weeks it was 2.6-2.8 in all groups (n ≥ 9/group). MSCs accelerated SF onset. At eight weeks post-INR, sciatic function index (SFI), a measure of MF (0 = Normal; -100 = Nonfunctional) was -34 and -77 in MSC and vehicle groups, respectively (n ≥ 9); post-SNR it was -72 and -92 in MSC and vehicle groups, respectively. Long-term MF (24 weeks) was apparent in MSC treated INR (SFI -63) but not in SNR (SFI -100). Gastrocnemius muscle atrophy was significantly reduced (P < 0.05) in INR. Nerve histomorphometry revealed reduced axonal area (P < 0.01) but no difference in myelination (P > 0.05) in MSC treated INR compared to the naive contralateral nerve. CONCLUSION: MSC therapy in peripheral nerve damage appears to improve nerve regeneration, mitigate flexion-contractures, and promote limb functional recovery.

15.
Nat Commun ; 8(1): 1603, 2017 11 17.
Article in English | MEDLINE | ID: mdl-29150673

ABSTRACT

To better understand the roles of microRNAs in glial function, we used a conditional deletion of Dicer1 (Dicer-CKOMG) in retinal Müller glia (MG). Dicer1 deletion from the MG leads to an abnormal migration of the cells as early as 1 month after the deletion. By 6 months after Dicer1 deletion, the MG form large aggregations and severely disrupt normal retinal architecture and function. The most highly upregulated gene in the Dicer-CKOMG MG is the proteoglycan Brevican (Bcan) and overexpression of Bcan results in similar aggregations of the MG in wild-type retina. One potential microRNA that regulates Bcan is miR-9, and overexpression of miR-9 can partly rescue the effects of Dicer1 deletion on the MG phenotype. We also find that MG from retinitis pigmentosa patients display an increase in Brevican immunoreactivity at sites of MG aggregation, linking the retinal remodeling that occurs in chronic disease with microRNAs.


Subject(s)
Ependymoglial Cells/metabolism , Homeostasis/genetics , MicroRNAs/genetics , Neuroglia/metabolism , Retina/metabolism , 3T3 Cells , Animals , Cell Movement/genetics , Cells, Cultured , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Gene Expression Profiling , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Neuroglia/cytology , Retina/cytology , Ribonuclease III/deficiency , Ribonuclease III/genetics
16.
Elife ; 62017 11 09.
Article in English | MEDLINE | ID: mdl-29120328

ABSTRACT

As more people live longer, age-related neurodegenerative diseases are an increasingly important societal health issue. Treatments targeting specific pathologies such as amyloid beta in Alzheimer's disease (AD) have not led to effective treatments, and there is increasing evidence of a disconnect between traditional pathology and cognitive abilities with advancing age, indicative of individual variation in resilience to pathology. Here, we generated a comprehensive neuropathological, molecular, and transcriptomic characterization of hippocampus and two regions cortex in 107 aged donors (median = 90) from the Adult Changes in Thought (ACT) study as a freely-available resource (http://aging.brain-map.org/). We confirm established associations between AD pathology and dementia, albeit with increased, presumably aging-related variability, and identify sets of co-expressed genes correlated with pathological tau and inflammation markers. Finally, we demonstrate a relationship between dementia and RNA quality, and find common gene signatures, highlighting the importance of properly controlling for RNA quality when studying dementia.


Subject(s)
Aging/pathology , Cerebral Cortex/pathology , Gene Expression Profiling , Hippocampus/pathology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Dementia/pathology , Female , Humans , Male
17.
Nature ; 548(7665): 103-107, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28746305

ABSTRACT

Many retinal diseases lead to the loss of retinal neurons and cause visual impairment. The adult mammalian retina has little capacity for regeneration. By contrast, teleost fish functionally regenerate their retina following injury, and Müller glia (MG) are the source of regenerated neurons. The proneural transcription factor Ascl1 is upregulated in MG after retinal damage in zebrafish and is necessary for regeneration. Although Ascl1 is not expressed in mammalian MG after injury, forced expression of Ascl1 in mouse MG induces a neurogenic state in vitro and in vivo after NMDA (N-methyl-d-aspartate) damage in young mice. However, by postnatal day 16, mouse MG lose neurogenic capacity, despite Ascl1 overexpression. Loss of neurogenic capacity in mature MG is accompanied by reduced chromatin accessibility, suggesting that epigenetic factors limit regeneration. Here we show that MG-specific overexpression of Ascl1, together with a histone deacetylase inhibitor, enables adult mice to generate neurons from MG after retinal injury. The MG-derived neurons express markers of inner retinal neurons, synapse with host retinal neurons, and respond to light. Using an assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq), we show that the histone deacetylase inhibitor promotes accessibility at key gene loci in the MG, and allows more effective reprogramming. Our results thus provide a new approach for the treatment of blinding retinal diseases.


Subject(s)
Nerve Regeneration , Neurogenesis , Neuroglia/cytology , Neurons/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Epistasis, Genetic/drug effects , Female , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Male , Mice , Nerve Regeneration/drug effects , Neural Pathways/drug effects , Neurogenesis/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Retina/cytology , Retina/metabolism , Synapses/drug effects , Synapses/metabolism
20.
J Neurosci Res ; 93(9): 1413-22, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25824456

ABSTRACT

Parkinson's disease and Alzheimer's disease (AD) are recognized to coexist on a spectrum of neurodegeneration, and it has been proposed that molecular interactions among pathogenic proteins are a basis for the overlap between these two diseases. We instead hypothesized that degeneration of the nigrostriatal dopaminergic system enhances the clinical penetrance of early-stage AD. To determine the effect of striatal dopamine (DA) on the pathological effects in an experimental model of AD, APPSWE /PS1ΔE9 mice received striatal injections of the neurotoxin 6-hydroxydopamine (6OHDA). Animals were tested in a Barnes maze protocol and in a water T-maze protocol at different ages to determine the onset of cognitive impairment. APPSWE /PS1ΔE9 mice that received 6OHDA injections showed significant impairment in Barnes maze performance at an earlier age than controls. Additionally, at 12 months of age, APPswe /PS1ΔE9 + 6OHDA mice demonstrated worse behavioral flexibility than other groups in a task-switch phase of the water T-maze. To determine the neuroprotective effects of dopaminergic neurotransmission against amyloid-ß42 (Aß42 ) toxicity, neuronal branch order and dendrite length were quantified in primary medium spiny neuron (MSN) cultures pretreated with increasing doses of the D1 and D2 receptor agonists before being exposed to oligomerized Aß42 . Although there were no differences in Aß peptide levels or plaque burden among the groups, in murine MSN culture dopaminergic agonists prevented a toxic response to Aß42. Depletion of DA in the striatum exacerbated the cognitive impairment seen in a mouse model of early-stage AD; this may be due to a protective effect of dopaminergic innervation against Aß striatal neurotoxicity.


Subject(s)
Alzheimer Disease/complications , Cognition Disorders/genetics , Corpus Striatum/metabolism , Dopamine/deficiency , Penetrance , Adrenergic Agents/toxicity , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Cognition Disorders/chemically induced , Cognition Disorders/pathology , Corpus Striatum/drug effects , Disease Models, Animal , Humans , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Mutation/genetics , Oxidopamine/toxicity , Presenilin-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...