Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(20): 3753-3769.e18, 2022 09 29.
Article in English | MEDLINE | ID: mdl-36179668

ABSTRACT

Interactions between angiogenesis and neurogenesis regulate embryonic brain development. However, a comprehensive understanding of the stages of vascular cell maturation is lacking, especially in the prenatal human brain. Using fluorescence-activated cell sorting, single-cell transcriptomics, and histological and ultrastructural analyses, we show that an ensemble of endothelial and mural cell subtypes tile the brain vasculature during the second trimester. These vascular cells follow distinct developmental trajectories and utilize diverse signaling mechanisms, including collagen, laminin, and midkine, to facilitate cell-cell communication and maturation. Interestingly, our results reveal that tip cells, a subtype of endothelial cells, are highly enriched near the ventricular zone, the site of active neurogenesis. Consistent with these observations, prenatal vascular cells transplanted into cortical organoids exhibit restricted lineage potential that favors tip cells, promotes neurogenesis, and reduces cellular stress. Together, our results uncover important mechanisms into vascular maturation during this critical period of human brain development.


Subject(s)
Endothelial Cells , Neovascularization, Physiologic , Brain , Collagen , Humans , Laminin , Midkine , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/physiology , Pericytes
2.
Cell ; 182(3): 594-608.e11, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32679030

ABSTRACT

Human cerebral cortex size and complexity has increased greatly during evolution. While increased progenitor diversity and enhanced proliferative potential play important roles in human neurogenesis and gray matter expansion, the mechanisms of human oligodendrogenesis and white matter expansion remain largely unknown. Here, we identify EGFR-expressing "Pre-OPCs" that originate from outer radial glial cells (oRGs) and undergo mitotic somal translocation (MST) during division. oRG-derived Pre-OPCs provide an additional source of human cortical oligodendrocyte precursor cells (OPCs) and define a lineage trajectory. We further show that human OPCs undergo consecutive symmetric divisions to exponentially increase the progenitor pool size. Additionally, we find that the OPC-enriched gene, PCDH15, mediates daughter cell repulsion and facilitates proliferation. These findings indicate properties of OPC derivation, proliferation, and dispersion important for human white matter expansion and myelination.


Subject(s)
Cadherins/metabolism , Cerebral Cortex/cytology , Ependymoglial Cells/metabolism , Neurogenesis/genetics , Oligodendrocyte Precursor Cells/metabolism , Cadherin Related Proteins , Cadherins/genetics , Cell Proliferation/genetics , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Ependymoglial Cells/cytology , ErbB Receptors/genetics , ErbB Receptors/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Oligodendrocyte Precursor Cells/cytology , RNA, Small Interfering , RNA-Seq , Single-Cell Analysis , White Matter/cytology , White Matter/embryology , White Matter/metabolism
3.
Cell ; 183(1): 126-142.e17, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32961131

ABSTRACT

CD19-directed immunotherapies are clinically effective for treating B cell malignancies but also cause a high incidence of neurotoxicity. A subset of patients treated with chimeric antigen receptor (CAR) T cells or bispecific T cell engager (BiTE) antibodies display severe neurotoxicity, including fatal cerebral edema associated with T cell infiltration into the brain. Here, we report that mural cells, which surround the endothelium and are critical for blood-brain-barrier integrity, express CD19. We identify CD19 expression in brain mural cells using single-cell RNA sequencing data and confirm perivascular staining at the protein level. CD19 expression in the brain begins early in development alongside the emergence of mural cell lineages and persists throughout adulthood across brain regions. Mouse mural cells demonstrate lower levels of Cd19 expression, suggesting limitations in preclinical animal models of neurotoxicity. These data suggest an on-target mechanism for neurotoxicity in CD19-directed therapies and highlight the utility of human single-cell atlases for designing immunotherapies.


Subject(s)
Blood-Brain Barrier/metabolism , Epithelial Cells/metabolism , Immunotherapy, Adoptive/adverse effects , Animals , Antibodies, Bispecific/immunology , Antigens, CD19/immunology , B-Lymphocytes/immunology , Blood-Brain Barrier/immunology , Brain/immunology , Brain/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred NOD , Mice, SCID , Muscle, Smooth, Vascular/metabolism , Neoplasms , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Single-Cell Analysis/methods , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
4.
Cell ; 182(2): 497-514.e22, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32579974

ABSTRACT

To define the cellular composition and architecture of cutaneous squamous cell carcinoma (cSCC), we combined single-cell RNA sequencing with spatial transcriptomics and multiplexed ion beam imaging from a series of human cSCCs and matched normal skin. cSCC exhibited four tumor subpopulations, three recapitulating normal epidermal states, and a tumor-specific keratinocyte (TSK) population unique to cancer, which localized to a fibrovascular niche. Integration of single-cell and spatial data mapped ligand-receptor networks to specific cell types, revealing TSK cells as a hub for intercellular communication. Multiple features of potential immunosuppression were observed, including T regulatory cell (Treg) co-localization with CD8 T cells in compartmentalized tumor stroma. Finally, single-cell characterization of human tumor xenografts and in vivo CRISPR screens identified essential roles for specific tumor subpopulation-enriched gene networks in tumorigenesis. These data define cSCC tumor and stromal cell subpopulations, the spatial niches where they interact, and the communicating gene networks that they engage in cancer.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Genomics/methods , Skin Neoplasms/metabolism , Animals , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , RNA-Seq , Single-Cell Analysis , Skin/metabolism , Skin Neoplasms/pathology , Transcriptome , Transplantation, Heterologous
5.
Cell ; 180(3): 568-584.e23, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31981491

ABSTRACT

We present the largest exome sequencing study of autism spectrum disorder (ASD) to date (n = 35,584 total samples, 11,986 with ASD). Using an enhanced analytical framework to integrate de novo and case-control rare variation, we identify 102 risk genes at a false discovery rate of 0.1 or less. Of these genes, 49 show higher frequencies of disruptive de novo variants in individuals ascertained to have severe neurodevelopmental delay, whereas 53 show higher frequencies in individuals ascertained to have ASD; comparing ASD cases with mutations in these groups reveals phenotypic differences. Expressed early in brain development, most risk genes have roles in regulation of gene expression or neuronal communication (i.e., mutations effect neurodevelopmental and neurophysiological changes), and 13 fall within loci recurrently hit by copy number variants. In cells from the human cortex, expression of risk genes is enriched in excitatory and inhibitory neuronal lineages, consistent with multiple paths to an excitatory-inhibitory imbalance underlying ASD.


Subject(s)
Autistic Disorder/genetics , Cerebral Cortex/growth & development , Exome Sequencing/methods , Gene Expression Regulation, Developmental , Neurobiology/methods , Case-Control Studies , Cell Lineage , Cohort Studies , Exome , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Mutation, Missense , Neurons/metabolism , Phenotype , Sex Factors , Single-Cell Analysis/methods
6.
Cell ; 176(4): 743-756.e17, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30735633

ABSTRACT

Direct comparisons of human and non-human primate brains can reveal molecular pathways underlying remarkable specializations of the human brain. However, chimpanzee tissue is inaccessible during neocortical neurogenesis when differences in brain size first appear. To identify human-specific features of cortical development, we leveraged recent innovations that permit generating pluripotent stem cell-derived cerebral organoids from chimpanzee. Despite metabolic differences, organoid models preserve gene regulatory networks related to primary cell types and developmental processes. We further identified 261 differentially expressed genes in human compared to both chimpanzee organoids and macaque cortex, enriched for recent gene duplications, and including multiple regulators of PI3K-AKT-mTOR signaling. We observed increased activation of this pathway in human radial glia, dependent on two receptors upregulated specifically in human: INSR and ITGB8. Our findings establish a platform for systematic analysis of molecular changes contributing to human brain development and evolution.


Subject(s)
Cerebral Cortex/cytology , Organoids/metabolism , Animals , Biological Evolution , Brain/cytology , Cell Culture Techniques/methods , Cell Differentiation/genetics , Cerebral Cortex/metabolism , Gene Regulatory Networks/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Macaca , Neurogenesis/genetics , Organoids/growth & development , Pan troglodytes , Pluripotent Stem Cells/cytology , Single-Cell Analysis , Species Specificity , Transcriptome/genetics
7.
Cell ; 173(6): 1356-1369.e22, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29856954

ABSTRACT

Genetic changes causing brain size expansion in human evolution have remained elusive. Notch signaling is essential for radial glia stem cell proliferation and is a determinant of neuronal number in the mammalian cortex. We find that three paralogs of human-specific NOTCH2NL are highly expressed in radial glia. Functional analysis reveals that different alleles of NOTCH2NL have varying potencies to enhance Notch signaling by interacting directly with NOTCH receptors. Consistent with a role in Notch signaling, NOTCH2NL ectopic expression delays differentiation of neuronal progenitors, while deletion accelerates differentiation into cortical neurons. Furthermore, NOTCH2NL genes provide the breakpoints in 1q21.1 distal deletion/duplication syndrome, where duplications are associated with macrocephaly and autism and deletions with microcephaly and schizophrenia. Thus, the emergence of human-specific NOTCH2NL genes may have contributed to the rapid evolution of the larger human neocortex, accompanied by loss of genomic stability at the 1q21.1 locus and resulting recurrent neurodevelopmental disorders.


Subject(s)
Brain/embryology , Cerebral Cortex/physiology , Neurogenesis/physiology , Receptor, Notch2/metabolism , Signal Transduction , Animals , Cell Differentiation , Embryonic Stem Cells/metabolism , Female , Gene Deletion , Genes, Reporter , Gorilla gorilla , HEK293 Cells , Humans , Neocortex/cytology , Neural Stem Cells/metabolism , Neuroglia/metabolism , Neurons/metabolism , Pan troglodytes , Receptor, Notch2/genetics , Sequence Analysis, RNA
9.
Nature ; 623(7986): 274-282, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37938705

ABSTRACT

Neuroscience research has entered a phase of key discoveries in the realm of neurogenomics owing to strong financial and intellectual support for resource building and tool development. The previous challenge of tissue heterogeneity has been met with the application of techniques that can profile individual cells at scale. Moreover, the ability to perturb genes, gene regulatory elements and neuronal activity in a cell-type-specific manner has been integrated with gene expression studies to uncover the functional underpinnings of the genome at a systems level. Although these insights have necessarily been grounded in model systems, we now have the opportunity to apply these approaches in humans and in human tissue, thanks to advances in human genetics, brain imaging and tissue collection. We acknowledge that there will probably always be limits to the extent to which we can apply the genomic tools developed in model systems to human neuroscience; however, as we describe in this Perspective, the neuroscience field is now primed with an optimal foundation for tackling this ambitious challenge. The application of systems-level network analyses to these datasets will facilitate a deeper appreciation of human neurogenomics that cannot otherwise be achieved from directly observable phenomena.


Subject(s)
Genomics , Neurosciences , Systems Biology , Humans , Brain/metabolism , Genomics/trends , Models, Biological , Neurosciences/methods , Neurosciences/trends , Systems Biology/trends
10.
Nature ; 598(7879): 200-204, 2021 10.
Article in English | MEDLINE | ID: mdl-34616070

ABSTRACT

The human brain is subdivided into distinct anatomical structures, including the neocortex, which in turn encompasses dozens of distinct specialized cortical areas. Early morphogenetic gradients are known to establish early brain regions and cortical areas, but how early patterns result in finer and more discrete spatial differences remains poorly understood1. Here we use single-cell RNA sequencing to profile ten major brain structures and six neocortical areas during peak neurogenesis and early gliogenesis. Within the neocortex, we find that early in the second trimester, a large number of genes are differentially expressed across distinct cortical areas in all cell types, including radial glia, the neural progenitors of the cortex. However, the abundance of areal transcriptomic signatures increases as radial glia differentiate into intermediate progenitor cells and ultimately give rise to excitatory neurons. Using an automated, multiplexed single-molecule fluorescent in situ hybridization approach, we find that laminar gene-expression patterns are highly dynamic across cortical regions. Together, our data suggest that early cortical areal patterning is defined by strong, mutually exclusive frontal and occipital gene-expression signatures, with resulting gradients giving rise to the specification of areas between these two poles throughout successive developmental timepoints.


Subject(s)
Gene Expression Regulation, Developmental , Neocortex/cytology , Neocortex/embryology , Atlases as Topic , Base Sequence , Biomarkers/metabolism , Humans , Neocortex/metabolism , Neurogenesis , Neuroglia/classification , Neuroglia/cytology , Neuroglia/metabolism , Neurons/classification , Neurons/cytology , Neurons/metabolism , Reproducibility of Results , Single-Cell Analysis , Time Factors
11.
Mol Cell ; 73(4): 830-844.e12, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30639242

ABSTRACT

Proximity-dependent biotin labeling (BioID) may identify new targets for cancers driven by difficult-to-drug oncogenes such as Ras. Therefore, BioID was used with wild-type (WT) and oncogenic mutant (MT) H-, K-, and N-Ras, identifying known interactors, including Raf and PI3K, as well as a common set of 130 novel proteins proximal to all Ras isoforms. A CRISPR screen of these proteins for Ras dependence identified mTOR, which was also found proximal to MT Ras in human tumors. Oncogenic Ras directly bound two mTOR complex 2 (mTORC2) components, mTOR and MAPKAP1, to promote mTORC2 kinase activity at the plasma membrane. mTORC2 enabled the Ras pro-proliferative cell cycle transcriptional program, and perturbing the Ras-mTORC2 interaction impaired Ras-dependent neoplasia in vivo. Combining proximity-dependent proteomics with CRISPR screening identified a new set of functional Ras-associated proteins, defined mTORC2 as a new direct Ras effector, and offers a strategy for finding new proteins that cooperate with dominant oncogenes.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Neoplasms/metabolism , Proteome , ras Proteins/metabolism , Animals , Binding Sites , CRISPR-Cas Systems , Caco-2 Cells , Cell Cycle Checkpoints , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 2/genetics , Mice, Hairless , Mice, SCID , Mice, Transgenic , Mutation , Neoplasms/genetics , Neoplasms/pathology , Protein Binding , Protein Interaction Domains and Motifs , Proteomics/methods , Tumor Burden , ras Proteins/genetics
12.
Nature ; 578(7793): 142-148, 2020 02.
Article in English | MEDLINE | ID: mdl-31996853

ABSTRACT

Cortical organoids are self-organizing three-dimensional cultures that model features of the developing human cerebral cortex1,2. However, the fidelity of organoid models remains unclear3-5. Here we analyse the transcriptomes of individual primary human cortical cells from different developmental periods and cortical areas. We find that cortical development is characterized by progenitor maturation trajectories, the emergence of diverse cell subtypes and areal specification of newborn neurons. By contrast, organoids contain broad cell classes, but do not recapitulate distinct cellular subtype identities and appropriate progenitor maturation. Although the molecular signatures of cortical areas emerge in organoid neurons, they are not spatially segregated. Organoids also ectopically activate cellular stress pathways, which impairs cell-type specification. However, organoid stress and subtype defects are alleviated by transplantation into the mouse cortex. Together, these datasets and analytical tools provide a framework for evaluating and improving the accuracy of cortical organoids as models of human brain development.


Subject(s)
Cerebral Cortex , Neurogenesis , Stress, Physiological , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Humans , Neurons , Organoids , Single-Cell Analysis , Tissue Culture Techniques
14.
Nature ; 573(7772): 75-82, 2019 09.
Article in English | MEDLINE | ID: mdl-31316211

ABSTRACT

Multiple sclerosis (MS) is a neuroinflammatory disease with a relapsing-remitting disease course at early stages, distinct lesion characteristics in cortical grey versus subcortical white matter and neurodegeneration at chronic stages. Here we used single-nucleus RNA sequencing to assess changes in expression in multiple cell lineages in MS lesions and validated the results using multiplex in situ hybridization. We found selective vulnerability and loss of excitatory CUX2-expressing projection neurons in upper-cortical layers underlying meningeal inflammation; such MS neuron populations exhibited upregulation of stress pathway genes and long non-coding RNAs. Signatures of stressed oligodendrocytes, reactive astrocytes and activated microglia mapped most strongly to the rim of MS plaques. Notably, single-nucleus RNA sequencing identified phagocytosing microglia and/or macrophages by their ingestion and perinuclear import of myelin transcripts, confirmed by functional mouse and human culture assays. Our findings indicate lineage- and region-specific transcriptomic changes associated with selective cortical neuron damage and glial activation contributing to progression of MS lesions.


Subject(s)
Cell Lineage , Multiple Sclerosis/pathology , Neurons/pathology , Adult , Animals , Astrocytes/metabolism , Astrocytes/pathology , Autopsy , Cryopreservation , Female , Homeodomain Proteins/metabolism , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Microglia/metabolism , Microglia/pathology , Middle Aged , Multiple Sclerosis/genetics , Myelin Sheath/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Phagocytosis , RNA, Small Nuclear/analysis , RNA, Small Nuclear/genetics , RNA-Seq , Transcriptome/genetics
15.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Article in English | MEDLINE | ID: mdl-34155100

ABSTRACT

Studies of the spatiotemporal, transcriptomic, and morphological diversity of radial glia (RG) have spurred our current models of human corticogenesis. In the developing cortex, neural intermediate progenitor cells (nIPCs) are a neuron-producing transit-amplifying cell type born in the germinal zones of the cortex from RG. The potential diversity of the nIPC population, that produces a significant portion of excitatory cortical neurons, is understudied, particularly in the developing human brain. Here we explore the spatiotemporal, transcriptomic, and morphological variation that exists within the human nIPC population and provide a resource for future studies. We observe that the spatial distribution of nIPCs in the cortex changes abruptly around gestational week (GW) 19/20, marking a distinct shift in cellular distribution and organization during late neurogenesis. We also identify five transcriptomic subtypes, one of which appears at this spatiotemporal transition. Finally, we observe a diversity of nIPC morphologies that do not correlate with specific transcriptomic subtypes. These results provide an analysis of the spatiotemporal, transcriptional, and morphological diversity of nIPCs in developing brain tissue and provide an atlas of nIPC subtypes in the developing human cortex that can benchmark in vitro models of human development such as cerebral organoids and help inform future studies of how nIPCs contribute to cortical neurogenesis.


Subject(s)
Cerebral Cortex/embryology , Neural Stem Cells/cytology , Green Fluorescent Proteins/metabolism , Humans , Neural Stem Cells/metabolism , Neurogenesis , Reproducibility of Results , Sequence Analysis, RNA , Single-Cell Analysis , Time Factors , Transcriptome/genetics
16.
Bioinformatics ; 37(23): 4578-4580, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34244710

ABSTRACT

SUMMARY: As the use of single-cell technologies has grown, so has the need for tools to explore these large, complicated datasets. The UCSC Cell Browser is a tool that allows scientists to visualize gene expression and metadata annotation distribution throughout a single-cell dataset or multiple datasets. AVAILABILITY AND IMPLEMENTATION: We provide the UCSC Cell Browser as a free website where scientists can explore a growing collection of single-cell datasets and a freely available python package for scientists to create stable, self-contained visualizations for their own single-cell datasets. Learn more at https://cells.ucsc.edu. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genomics , Software , Databases, Genetic , Metadata
17.
J Anat ; 235(3): 543-554, 2019 09.
Article in English | MEDLINE | ID: mdl-30644551

ABSTRACT

Neuroserpin is a serine-protease inhibitor mainly expressed in the CNS and involved in the inhibition of the proteolytic cascade. Animal models confirmed its neuroprotective role in perinatal hypoxia-ischaemia and adult stroke. Although neuroserpin may be a potential therapeutic target in the treatment of the aforementioned conditions, there is still no information in the literature on its distribution during human brain development. The present study provides a detailed description of the changing spatiotemporal patterns of neuroserpin focusing on physiological human brain development. Five stages were distinguished within our examined age range which spanned from the 7th gestational week until adulthood. In particular, subplate and deep cortical plate neurons were identified as the main sources of neuroserpin production between the 25th gestational week and the first postnatal month. Our immunohistochemical findings were substantiated by single cell RNA sequencing data showing specific neuronal and glial cell types expressing neuroserpin. The characterization of neuroserpin expression during physiological human brain development is essential for forthcoming studies which will explore its involvement in pathological conditions, such as perinatal hypoxia-ischaemia and adult stroke in human.


Subject(s)
Brain/embryology , Neuropeptides/metabolism , Serpins/metabolism , Brain/metabolism , Humans , Immunohistochemistry , Sequence Analysis, RNA , Single-Cell Analysis , Neuroserpin
18.
Nature ; 563(7729): 38-39, 2018 11.
Article in English | MEDLINE | ID: mdl-30382219

Subject(s)
Neocortex , Transcriptome , Brain
19.
BMC Biol ; 16(1): 113, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30309354

ABSTRACT

BACKGROUND: High throughput methods for profiling the transcriptomes of single cells have recently emerged as transformative approaches for large-scale population surveys of cellular diversity in heterogeneous primary tissues. However, the efficient generation of such atlases will depend on sufficient sampling of diverse cell types while remaining cost-effective to enable a comprehensive examination of organs, developmental stages, and individuals. RESULTS: To examine the relationship between sampled cell numbers and transcriptional heterogeneity in the context of unbiased cell type classification, we explored the population structure of a publicly available 1.3 million cell dataset from E18.5 mouse brain and validated our findings in published data from adult mice. We propose a computational framework for inferring the saturation point of cluster discovery in a single-cell mRNA-seq experiment, centered around cluster preservation in downsampled datasets. In addition, we introduce a "complexity index," which characterizes the heterogeneity of cells in a given dataset. Using Cajal-Retzius cells as an example of a limited complexity dataset, we explored whether the detected biological distinctions relate to technical clustering. Surprisingly, we found that clustering distinctions carrying biologically interpretable meaning are achieved with far fewer cells than the originally sampled, though technical saturation of rare populations such as Cajal-Retzius cells is not achieved. We additionally validated these findings with a recently published atlas of cell types across mouse organs and again find using subsampling that a much smaller number of cells recapitulates the cluster distinctions of the complete dataset. CONCLUSIONS: Together, these findings suggest that most of the biologically interpretable cell types from the 1.3 million cell database can be recapitulated by analyzing 50,000 randomly selected cells, indicating that instead of profiling few individuals at high "cellular coverage," cell atlas studies may instead benefit from profiling more individuals, or many time points at lower cellular coverage and then further enriching for populations of interest. This strategy is ideal for scenarios where cost and time are limited, though extremely rare populations of interest (< 1%) may be identifiable only with much higher cell numbers.


Subject(s)
Brain/physiology , Gene Expression Profiling/methods , High-Throughput Screening Assays/methods , Single-Cell Analysis/methods , Animals , Mice , Sampling Studies
SELECTION OF CITATIONS
SEARCH DETAIL