Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Cell ; 184(12): 3222-3241.e26, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34004146

ABSTRACT

The isocortex and hippocampal formation (HPF) in the mammalian brain play critical roles in perception, cognition, emotion, and learning. We profiled ∼1.3 million cells covering the entire adult mouse isocortex and HPF and derived a transcriptomic cell-type taxonomy revealing a comprehensive repertoire of glutamatergic and GABAergic neuron types. Contrary to the traditional view of HPF as having a simpler cellular organization, we discover a complete set of glutamatergic types in HPF homologous to all major subclasses found in the six-layered isocortex, suggesting that HPF and the isocortex share a common circuit organization. We also identify large-scale continuous and graded variations of cell types along isocortical depth, across the isocortical sheet, and in multiple dimensions in hippocampus and subiculum. Overall, our study establishes a molecular architecture of the mammalian isocortex and hippocampal formation and begins to shed light on its underlying relationship with the development, evolution, connectivity, and function of these two brain structures.


Subject(s)
Hippocampus/cytology , Neocortex/cytology , Transcriptome/genetics , Animals , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Mice, Inbred C57BL , Mice, Transgenic
2.
Cell ; 183(4): 935-953.e19, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33186530

ABSTRACT

Neurons are frequently classified into distinct types on the basis of structural, physiological, or genetic attributes. To better constrain the definition of neuronal cell types, we characterized the transcriptomes and intrinsic physiological properties of over 4,200 mouse visual cortical GABAergic interneurons and reconstructed the local morphologies of 517 of those neurons. We find that most transcriptomic types (t-types) occupy specific laminar positions within visual cortex, and, for most types, the cells mapping to a t-type exhibit consistent electrophysiological and morphological properties. These properties display both discrete and continuous variation among t-types. Through multimodal integrated analysis, we define 28 met-types that have congruent morphological, electrophysiological, and transcriptomic properties and robust mutual predictability. We identify layer-specific axon innervation pattern as a defining feature distinguishing different met-types. These met-types represent a unified definition of cortical GABAergic interneuron types, providing a systematic framework to capture existing knowledge and bridge future analyses across different modalities.


Subject(s)
Cerebral Cortex/cytology , Electrophysiological Phenomena , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Transcriptome/genetics , Animals , Female , Gene Expression Profiling , Hippocampus/physiology , Ion Channels/metabolism , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism
3.
Cell ; 149(2): 483-96, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22500809

ABSTRACT

Although there have been major advances in elucidating the functional biology of the human brain, relatively little is known of its cellular and molecular organization. Here we report a large-scale characterization of the expression of ∼1,000 genes important for neural functions by in situ hybridization at a cellular resolution in visual and temporal cortices of adult human brains. These data reveal diverse gene expression patterns and remarkable conservation of each individual gene's expression among individuals (95%), cortical areas (84%), and between human and mouse (79%). A small but substantial number of genes (21%) exhibited species-differential expression. Distinct molecular signatures, comprised of genes both common between species and unique to each, were identified for each major cortical cell type. The data suggest that gene expression profile changes may contribute to differential cortical function across species, and in particular, a shift from corticosubcortical to more predominant corticocortical communications in the human brain.


Subject(s)
Gene Expression Profiling , Neocortex/metabolism , Temporal Lobe/metabolism , Visual Cortex/metabolism , Adult , Animals , Gene Expression Regulation , Humans , Mice , Neocortex/cytology , Neurons/metabolism , Species Specificity , Temporal Lobe/cytology , Visual Cortex/cytology
4.
Proc Natl Acad Sci U S A ; 119(15): e2108760119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377797

ABSTRACT

Enhancers integrate transcription factor signaling pathways that drive cell fate specification in the developing brain. We paired enhancer labeling and single-cell RNA-sequencing (scRNA-seq) to delineate and distinguish specification of neuronal lineages in mouse medial, lateral, and caudal ganglionic eminences (MGE, LGE, and CGE) at embryonic day (E)11.5. We show that scRNA-seq clustering using transcription factors improves resolution of regional and developmental populations, and that enhancer activities identify specific and overlapping GE-derived neuronal populations. First, we mapped the activities of seven evolutionarily conserved brain enhancers at single-cell resolution in vivo, finding that the selected enhancers had diverse activities in specific progenitor and neuronal populations across the GEs. We then applied enhancer-based labeling, scRNA-seq, and analysis of in situ hybridization data to distinguish transcriptionally distinct and spatially defined subtypes of MGE-derived GABAergic and cholinergic projection neurons and interneurons. Our results map developmental origins and specification paths underlying neurogenesis in the embryonic basal ganglia and showcase the power of scRNA-seq combined with enhancer-based labeling to resolve the complex paths of neuronal specification underlying mouse brain development.


Subject(s)
Basal Ganglia , Cholinergic Neurons , Enhancer Elements, Genetic , GABAergic Neurons , Neurogenesis , Animals , Basal Ganglia/cytology , Basal Ganglia/embryology , Cell Lineage/genetics , Cholinergic Neurons/metabolism , GABAergic Neurons/metabolism , Mice , Neurogenesis/genetics , RNA-Seq , Single-Cell Analysis , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nature ; 508(7495): 207-14, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24695228

ABSTRACT

Comprehensive knowledge of the brain's wiring diagram is fundamental for understanding how the nervous system processes information at both local and global scales. However, with the singular exception of the C. elegans microscale connectome, there are no complete connectivity data sets in other species. Here we report a brain-wide, cellular-level, mesoscale connectome for the mouse. The Allen Mouse Brain Connectivity Atlas uses enhanced green fluorescent protein (EGFP)-expressing adeno-associated viral vectors to trace axonal projections from defined regions and cell types, and high-throughput serial two-photon tomography to image the EGFP-labelled axons throughout the brain. This systematic and standardized approach allows spatial registration of individual experiments into a common three dimensional (3D) reference space, resulting in a whole-brain connectivity matrix. A computational model yields insights into connectional strength distribution, symmetry and other network properties. Virtual tractography illustrates 3D topography among interconnected regions. Cortico-thalamic pathway analysis demonstrates segregation and integration of parallel pathways. The Allen Mouse Brain Connectivity Atlas is a freely available, foundational resource for structural and functional investigations into the neural circuits that support behavioural and cognitive processes in health and disease.


Subject(s)
Brain/anatomy & histology , Brain/cytology , Connectome , Animals , Atlases as Topic , Axons/physiology , Cerebral Cortex/cytology , Corpus Striatum/cytology , Male , Mice , Mice, Inbred C57BL , Models, Neurological , Neuroanatomical Tract-Tracing Techniques , Thalamus/cytology
6.
Nature ; 489(7416): 391-399, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22996553

ABSTRACT

Neuroanatomically precise, genome-wide maps of transcript distributions are critical resources to complement genomic sequence data and to correlate functional and genetic brain architecture. Here we describe the generation and analysis of a transcriptional atlas of the adult human brain, comprising extensive histological analysis and comprehensive microarray profiling of ∼900 neuroanatomically precise subdivisions in two individuals. Transcriptional regulation varies enormously by anatomical location, with different regions and their constituent cell types displaying robust molecular signatures that are highly conserved between individuals. Analysis of differential gene expression and gene co-expression relationships demonstrates that brain-wide variation strongly reflects the distributions of major cell classes such as neurons, oligodendrocytes, astrocytes and microglia. Local neighbourhood relationships between fine anatomical subdivisions are associated with discrete neuronal subtypes and genes involved with synaptic transmission. The neocortex displays a relatively homogeneous transcriptional pattern, but with distinct features associated selectively with primary sensorimotor cortices and with enriched frontal lobe expression. Notably, the spatial topography of the neocortex is strongly reflected in its molecular topography-the closer two cortical regions, the more similar their transcriptomes. This freely accessible online data resource forms a high-resolution transcriptional baseline for neurogenetic studies of normal and abnormal human brain function.


Subject(s)
Anatomy, Artistic , Atlases as Topic , Brain/anatomy & histology , Brain/metabolism , Gene Expression Profiling , Transcriptome/genetics , Adult , Animals , Brain/cytology , Calbindins , Databases, Genetic , Dopamine/metabolism , Health , Hippocampus/cytology , Hippocampus/metabolism , Humans , In Situ Hybridization , Internet , Macaca mulatta/anatomy & histology , Macaca mulatta/genetics , Male , Mice , Neocortex/anatomy & histology , Neocortex/cytology , Neocortex/metabolism , Oligonucleotide Array Sequence Analysis , Post-Synaptic Density/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , S100 Calcium Binding Protein G/genetics , Species Specificity
7.
Eur J Neurosci ; 45(8): 1013-1023, 2017 04.
Article in English | MEDLINE | ID: mdl-28177156

ABSTRACT

Despite the widespread use of current-source density (CSD) analysis of extracellular potential recordings in the brain, the physical mechanisms responsible for the generation of the signal are still debated. While the extracellular potential is thought to be exclusively generated by the transmembrane currents, recent studies suggest that extracellular diffusive, advective and displacement currents-traditionally neglected-may also contribute considerably toward extracellular potential recordings. Here, we first justify the application of the electro-quasistatic approximation of Maxwell's equations to describe the electromagnetic field of physiological origin. Subsequently, we perform spatial averaging of currents in neural tissue to arrive at the notion of the CSD and derive an equation relating it to the extracellular potential. We show that, in general, the extracellular potential is determined by the CSD of membrane currents as well as the gradients of the putative extracellular diffusion current. The diffusion current can contribute significantly to the extracellular potential at frequencies less than a few Hertz; in which case it must be subtracted to obtain correct CSD estimates. We also show that the advective and displacement currents in the extracellular space are negligible for physiological frequencies while, within cellular membrane, displacement current contributes toward the CSD as a capacitive current. Taken together, these findings elucidate the relationship between electric currents and the extracellular potential in brain tissue and form the necessary foundation for the analysis of extracellular recordings.


Subject(s)
Brain/physiology , Membrane Potentials/physiology , Models, Neurological , Neurons/physiology , Algorithms , Animals , Diffusion , Electricity , Electrodes, Implanted , Electromagnetic Fields , Male , Mice, Inbred C57BL , Photic Stimulation , Synaptic Transmission/physiology , Visual Perception/physiology
8.
Development ; 140(22): 4633-44, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24154525

ABSTRACT

The neurogenic potential of the subgranular zone (SGZ) of the hippocampal dentate gyrus is likely to be regulated by molecular cues arising from its complex heterogeneous cellular environment. Through transcriptome analysis using laser microdissection coupled with DNA microarrays, in combination with analysis of genome-wide in situ hybridization data, we identified 363 genes selectively enriched in adult mouse SGZ. These genes reflect expression in the different constituent cell types, including progenitor and dividing cells, immature granule cells, astrocytes, oligodendrocytes and GABAergic interneurons. Similar transcriptional profiling in the rhesus monkey dentate gyrus across postnatal development identified a highly overlapping set of SGZ-enriched genes, which can be divided based on temporal profiles to reflect maturation of glia versus granule neurons. Furthermore, we identified a neurogenesis-related gene network with decreasing postnatal expression that is highly correlated with the declining number of proliferating cells in dentate gyrus over postnatal development. Many of the genes in this network showed similar postnatal downregulation in mouse, suggesting a conservation of molecular mechanisms underlying developmental and adult neurogenesis in rodents and primates. Conditional deletion of Sox4 and Sox11, encoding two neurogenesis-related transcription factors central in this network, produces a mouse with no hippocampus, confirming the crucial role for these genes in regulating hippocampal neurogenesis.


Subject(s)
Gene Expression Profiling , Hippocampus/metabolism , Macaca mulatta/genetics , Neurogenesis/genetics , Animals , Animals, Newborn , Biomarkers/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genome/genetics , Hippocampus/cytology , Interneurons/cytology , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Multigene Family , Oligodendroglia/cytology , Oligodendroglia/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Spatio-Temporal Analysis , Transcription, Genetic
9.
Methods ; 73: 79-89, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25449901

ABSTRACT

The Allen Brain Atlases enable the study of spatially resolved, genome-wide gene expression patterns across the mammalian brain. Several explorative studies have applied linear dimensionality reduction methods such as Principal Component Analysis (PCA) and classical Multi-Dimensional Scaling (cMDS) to gain insight into the spatial organization of these expression patterns. In this paper, we describe a non-linear embedding technique called Barnes-Hut Stochastic Neighbor Embedding (BH-SNE) that emphasizes the local similarity structure of high-dimensional data points. By applying BH-SNE to the gene expression data from the Allen Brain Atlases, we demonstrate the consistency of the 2D, non-linear embedding of the sagittal and coronal mouse brain atlases, and across 6 human brains. In addition, we quantitatively show that BH-SNE maps are superior in their separation of neuroanatomical regions in comparison to PCA and cMDS. Finally, we assess the effect of higher-order principal components on the global structure of the BH-SNE similarity maps. Based on our observations, we conclude that BH-SNE maps with or without prior dimensionality reduction (based on PCA) provide comprehensive and intuitive insights in both the local and global spatial transcriptome structure of the human and mouse Allen Brain Atlases.


Subject(s)
Atlases as Topic , Brain Mapping/methods , Brain/anatomy & histology , Brain/physiology , Nonlinear Dynamics , Transcriptome/genetics , Adult , Animals , Female , Gene Expression Regulation , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Young Adult
10.
Nature ; 445(7124): 168-76, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17151600

ABSTRACT

Molecular approaches to understanding the functional circuitry of the nervous system promise new insights into the relationship between genes, brain and behaviour. The cellular diversity of the brain necessitates a cellular resolution approach towards understanding the functional genomics of the nervous system. We describe here an anatomically comprehensive digital atlas containing the expression patterns of approximately 20,000 genes in the adult mouse brain. Data were generated using automated high-throughput procedures for in situ hybridization and data acquisition, and are publicly accessible online. Newly developed image-based informatics tools allow global genome-scale structural analysis and cross-correlation, as well as identification of regionally enriched genes. Unbiased fine-resolution analysis has identified highly specific cellular markers as well as extensive evidence of cellular heterogeneity not evident in classical neuroanatomical atlases. This highly standardized atlas provides an open, primary data resource for a wide variety of further studies concerning brain organization and function.


Subject(s)
Brain/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genome/genetics , Animals , Brain/anatomy & histology , Brain/cytology , Computational Biology , Genomics , Hippocampus/anatomy & histology , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Proc Natl Acad Sci U S A ; 107(44): 19049-54, 2010 Nov 02.
Article in English | MEDLINE | ID: mdl-20956311

ABSTRACT

Considerable progress has been made in understanding variations in gene sequence and expression level associated with phenotype, yet how genetic diversity translates into complex phenotypic differences remains poorly understood. Here, we examine the relationship between genetic background and spatial patterns of gene expression across seven strains of mice, providing the most extensive cellular-resolution comparative analysis of gene expression in the mammalian brain to date. Using comprehensive brainwide anatomic coverage (more than 200 brain regions), we applied in situ hybridization to analyze the spatial expression patterns of 49 genes encoding well-known pharmaceutical drug targets. Remarkably, over 50% of the genes examined showed interstrain expression variation. In addition, the variability was nonuniformly distributed across strain and neuroanatomic region, suggesting certain organizing principles. First, the degree of expression variance among strains mirrors genealogic relationships. Second, expression pattern differences were concentrated in higher-order brain regions such as the cortex and hippocampus. Divergence in gene expression patterns across the brain could contribute significantly to variations in behavior and responses to neuroactive drugs in laboratory mouse strains and may help to explain individual differences in human responsiveness to neuroactive drugs.


Subject(s)
Brain/metabolism , Gene Expression Regulation/physiology , Animals , Brain/cytology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , In Situ Hybridization , Mice , Species Specificity
12.
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
13.
BMC Genomics ; 13: 214, 2012 May 31.
Article in English | MEDLINE | ID: mdl-22651826

ABSTRACT

BACKGROUND: Post-transcriptional control of gene expression is mostly conducted by specific elements in untranslated regions (UTRs) of mRNAs, in collaboration with specific binding proteins and RNAs. In several well characterized cases, these RNA elements are known to form stable secondary structures. RNA secondary structures also may have major functional implications for long noncoding RNAs (lncRNAs). Recent transcriptional data has indicated the importance of lncRNAs in brain development and function. However, no methodical efforts to investigate this have been undertaken. Here, we aim to systematically analyze the potential for RNA structure in brain-expressed transcripts. RESULTS: By comprehensive spatial expression analysis of the adult mouse in situ hybridization data of the Allen Mouse Brain Atlas, we show that transcripts (coding as well as non-coding) associated with in silico predicted structured probes are highly and significantly enriched in almost all analyzed brain regions. Functional implications of these RNA structures and their role in the brain are discussed in detail along with specific examples. We observe that mRNAs with a structure prediction in their UTRs are enriched for binding, transport and localization gene ontology categories. In addition, after manual examination we observe agreement between RNA binding protein interaction sites near the 3' UTR structures and correlated expression patterns. CONCLUSIONS: Our results show a potential use for RNA structures in expressed coding as well as noncoding transcripts in the adult mouse brain, and describe the role of structured RNAs in the context of intracellular signaling pathways and regulatory networks. Based on this data we hypothesize that RNA structure is widely involved in transcriptional and translational regulatory mechanisms in the brain and ultimately plays a role in brain function.


Subject(s)
Brain/metabolism , Computational Biology/methods , Nucleic Acid Conformation , RNA/chemistry , RNA/genetics , Anatomy, Artistic , Animals , Atlases as Topic , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks/genetics , Genetic Variation , In Situ Hybridization , Mice , Molecular Sequence Annotation , Protein Binding/genetics , RNA Probes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Thermodynamics , Untranslated Regions/genetics
14.
Neuroinformatics ; 20(2): 507-512, 2022 04.
Article in English | MEDLINE | ID: mdl-35061216

ABSTRACT

In this perspective article, we consider the critical issue of data and other research object standardisation and, specifically, how international collaboration, and organizations such as the International Neuroinformatics Coordinating Facility (INCF) can encourage that emerging neuroscience data be Findable, Accessible, Interoperable, and Reusable (FAIR). As neuroscientists engaged in the sharing and integration of multi-modal and multiscale data, we see the current insufficiency of standards as a major impediment in the Interoperability and Reusability of research results. We call for increased international collaborative standardisation of neuroscience data to foster integration and efficient reuse of research objects.


Subject(s)
Data Collection , Neurosciences
15.
J Comp Neurol ; 530(1): 6-503, 2022 01.
Article in English | MEDLINE | ID: mdl-34525221

ABSTRACT

Increasing interest in studies of prenatal human brain development, particularly using new single-cell genomics and anatomical technologies to create cell atlases, creates a strong need for accurate and detailed anatomical reference atlases. In this study, we present two cellular-resolution digital anatomical atlases for prenatal human brain at postconceptional weeks (PCW) 15 and 21. Both atlases were annotated on sequential Nissl-stained sections covering brain-wide structures on the basis of combined analysis of cytoarchitecture, acetylcholinesterase staining, and an extensive marker gene expression dataset. This high information content dataset allowed reliable and accurate demarcation of developing cortical and subcortical structures and their subdivisions. Furthermore, using the anatomical atlases as a guide, spatial expression of 37 and 5 genes from the brains, respectively, at PCW 15 and 21 was annotated, illustrating reliable marker genes for many developing brain structures. Finally, the present study uncovered several novel developmental features, such as the lack of an outer subventricular zone in the hippocampal formation and entorhinal cortex, and the apparent extension of both cortical (excitatory) and subcortical (inhibitory) progenitors into the prenatal olfactory bulb. These comprehensive atlases provide useful tools for visualization, segmentation, targeting, imaging, and interpretation of brain structures of prenatal human brain, and for guiding and interpreting the next generation of cell census and connectome studies.


Subject(s)
Atlases as Topic , Brain/growth & development , Entorhinal Cortex/growth & development , Hippocampus/growth & development , Animals , Female , Humans , Pregnancy
16.
Elife ; 102021 09 02.
Article in English | MEDLINE | ID: mdl-34473054

ABSTRACT

Abundant evidence supports the presence of at least three distinct types of thalamocortical (TC) neurons in the primate dorsal lateral geniculate nucleus (dLGN) of the thalamus, the brain region that conveys visual information from the retina to the primary visual cortex (V1). Different types of TC neurons in mice, humans, and macaques have distinct morphologies, distinct connectivity patterns, and convey different aspects of visual information to the cortex. To investigate the molecular underpinnings of these cell types, and how these relate to differences in dLGN between human, macaque, and mice, we profiled gene expression in single nuclei and cells using RNA-sequencing. These efforts identified four distinct types of TC neurons in the primate dLGN: magnocellular (M) neurons, parvocellular (P) neurons, and two types of koniocellular (K) neurons. Despite extensively documented morphological and physiological differences between M and P neurons, we identified few genes with significant differential expression between transcriptomic cell types corresponding to these two neuronal populations. Likewise, the dominant feature of TC neurons of the adult mouse dLGN is high transcriptomic similarity, with an axis of heterogeneity that aligns with core vs. shell portions of mouse dLGN. Together, these data show that transcriptomic differences between principal cell types in the mature mammalian dLGN are subtle relative to the observed differences in morphology and cortical projection targets. Finally, alignment of transcriptome profiles across species highlights expanded diversity of GABAergic neurons in primate versus mouse dLGN and homologous types of TC neurons in primates that are distinct from TC neurons in mouse.


Subject(s)
Cell Nucleus/genetics , Geniculate Bodies/metabolism , Neurons/metabolism , Visual Cortex/metabolism , Animals , Gene Expression Profiling , Humans , Macaca , Mice , RNA-Seq , Single-Cell Analysis , Thalamus/metabolism , Visual Pathways/metabolism
17.
Elife ; 92020 12 29.
Article in English | MEDLINE | ID: mdl-33372656

ABSTRACT

The advancement of single-cell RNA-sequencing technologies has led to an explosion of cell type definitions across multiple organs and organisms. While standards for data and metadata intake are arising, organization of cell types has largely been left to individual investigators, resulting in widely varying nomenclature and limited alignment between taxonomies. To facilitate cross-dataset comparison, the Allen Institute created the common cell type nomenclature (CCN) for matching and tracking cell types across studies that is qualitatively similar to gene transcript management across different genome builds. The CCN can be readily applied to new or established taxonomies and was applied herein to diverse cell type datasets derived from multiple quantifiable modalities. The CCN facilitates assigning accurate yet flexible cell type names in the mammalian cortex as a step toward community-wide efforts to organize multi-source, data-driven information related to cell type taxonomies from any organism.


Subject(s)
Cells/classification , Temporal Lobe/cytology , Terminology as Topic , Humans
18.
Nat Commun ; 9(1): 710, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29459718

ABSTRACT

The cellular components of mammalian neocortical circuits are diverse, and capturing this diversity in computational models is challenging. Here we report an approach for generating biophysically detailed models of 170 individual neurons in the Allen Cell Types Database to link the systematic experimental characterization of cell types to the construction of cortical models. We build models from 3D morphologies and somatic electrophysiological responses measured in the same cells. Densities of active somatic conductances and additional parameters are optimized with a genetic algorithm to match electrophysiological features. We evaluate the models by applying additional stimuli and comparing model responses to experimental data. Applying this technique across a diverse set of neurons from adult mouse primary visual cortex, we verify that models preserve the distinctiveness of intrinsic properties between subsets of cells observed in experiments. The optimized models are accessible online alongside the experimental data. Code for optimization and simulation is also openly distributed.


Subject(s)
Neurons/physiology , Visual Cortex/cytology , Animals , Biophysics , Electrophysiological Phenomena , Mice , Models, Neurological , Neurons/chemistry , Visual Cortex/chemistry , Visual Cortex/physiology
19.
Brain Struct Funct ; 223(5): 2335-2342, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29478144

ABSTRACT

Decoding the chain from genes to cognition requires detailed insights how areas with specific gene activities and microanatomical architectures contribute to brain function and dysfunction. The Allen Human Brain Atlas contains regional gene expression data, while the JuBrain Atlas offers three-dimensional cytoarchitectonic maps reflecting interindividual variability. To date, an integrated framework that combines the analytical benefits of both scientific platforms towards a multi-level brain atlas of adult humans was not available. We have, therefore, developed JuGEx, a new method for integrating tissue transcriptome and cytoarchitectonic segregation. We investigated differential gene expression in two JuBrain areas of the frontal pole that we have structurally and functionally characterized in previous studies. Our results show a significant upregulation of MAOA and TAC1 in the medial area frontopolaris which is a node in the limbic-cortical network and known to be susceptible for gray matter loss and behavioral dysfunction in patients with depression. The MAOA gene encodes an enzyme which is involved in the catabolism of dopamine, norepinephrine, serotonin, and other monoaminergic neurotransmitters. The TAC1 locus generates hormones that play a role in neuron excitations and behavioral responses. Overall, JuGEx provides a new tool for the scientific community that empowers research from basic, cognitive and clinical neuroscience in brain regions and disease models with regard to gene expression.


Subject(s)
Brain Mapping , Frontal Lobe/anatomy & histology , Limbic System/anatomy & histology , Transcriptome/physiology , Adult , Aged , Aged, 80 and over , Analysis of Variance , Autopsy , Brain Mapping/methods , Female , Frontal Lobe/diagnostic imaging , Humans , Image Processing, Computer-Assisted , Limbic System/diagnostic imaging , Magnetic Resonance Imaging , Male , Middle Aged , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Neural Pathways/anatomy & histology , Oxygen/blood , Thyroid Nuclear Factor 1/genetics , Thyroid Nuclear Factor 1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL