Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Nature ; 575(7781): 195-202, 2019 11.
Article in English | MEDLINE | ID: mdl-31666704

ABSTRACT

The mammalian cortex is a laminar structure containing many areas and cell types that are densely interconnected in complex ways, and for which generalizable principles of organization remain mostly unknown. Here we describe a major expansion of the Allen Mouse Brain Connectivity Atlas resource1, involving around a thousand new tracer experiments in the cortex and its main satellite structure, the thalamus. We used Cre driver lines (mice expressing Cre recombinase) to comprehensively and selectively label brain-wide connections by layer and class of projection neuron. Through observations of axon termination patterns, we have derived a set of generalized anatomical rules to describe corticocortical, thalamocortical and corticothalamic projections. We have built a model to assign connection patterns between areas as either feedforward or feedback, and generated testable predictions of hierarchical positions for individual cortical and thalamic areas and for cortical network modules. Our results show that cell-class-specific connections are organized in a shallow hierarchy within the mouse corticothalamic network.


Subject(s)
Cerebral Cortex/anatomy & histology , Cerebral Cortex/cytology , Neural Pathways/anatomy & histology , Neural Pathways/cytology , Thalamus/anatomy & histology , Thalamus/cytology , Animals , Axons/physiology , Cerebral Cortex/physiology , Female , Integrases/genetics , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Thalamus/physiology
2.
Nat Methods ; 17(4): 422-429, 2020 04.
Article in English | MEDLINE | ID: mdl-32203389

ABSTRACT

Brain circuits comprise vast numbers of interconnected neurons with diverse molecular, anatomical and physiological properties. To allow targeting of individual neurons for structural and functional studies, we created light-inducible site-specific DNA recombinases based on Cre, Dre and Flp (RecVs). RecVs can induce genomic modifications by one-photon or two-photon light induction in vivo. They can produce targeted, sparse and strong labeling of individual neurons by modifying multiple loci within mouse and zebrafish genomes. In combination with other genetic strategies, they allow intersectional targeting of different neuronal classes. In the mouse cortex they enable sparse labeling and whole-brain morphological reconstructions of individual neurons. Furthermore, these enzymes allow single-cell two-photon targeted genetic modifications and can be used in combination with functional optical indicators with minimal interference. In summary, RecVs enable spatiotemporally precise optogenomic modifications that can facilitate detailed single-cell analysis of neural circuits by linking genetic identity, morphology, connectivity and function.


Subject(s)
Genomics/methods , Optogenetics , Recombinases/metabolism , Animals , Brain/cytology , Gene Expression Regulation , Genetic Engineering , Mice , Neurons/metabolism , Recombinases/genetics , Zebrafish
3.
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
4.
Cereb Cortex ; 25(2): 433-49, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24014670

ABSTRACT

The neocortex contains diverse populations of excitatory neurons segregated by layer and further definable by their specific cortical and subcortical projection targets. The current study describes a systematic approach to identify molecular correlates of specific projection neuron classes in mouse primary somatosensory cortex (S1), using a combination of in situ hybridization (ISH) data mining, marker gene colocalization, and combined retrograde labeling with ISH for layer-specific marker genes. First, we identified a large set of genes with specificity for each cortical layer, and that display heterogeneous patterns within those layers. Using these genes as markers, we find extensive evidence for the covariation of gene expression and projection target specificity in layer 2/3, 5, and 6, with individual genes labeling neurons projecting to specific subsets of target structures. The combination of gene expression and target specificity imply a great diversity of projection neuron classes that is similar to or greater than that of GABAergic interneurons. The covariance of these 2 phenotypic modalities suggests that these classes are both discrete and genetically specified.


Subject(s)
Neurons/cytology , Neurons/physiology , Somatosensory Cortex/cytology , Somatosensory Cortex/physiology , Animals , Atlases as Topic , Data Mining , Gene Expression/physiology , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Neural Pathways/cytology , Neural Pathways/physiology , Neuronal Tract-Tracers
5.
Nat Neurosci ; 26(2): 350-364, 2023 02.
Article in English | MEDLINE | ID: mdl-36550293

ABSTRACT

Identification of structural connections between neurons is a prerequisite to understanding brain function. Here we developed a pipeline to systematically map brain-wide monosynaptic input connections to genetically defined neuronal populations using an optimized rabies tracing system. We used mouse visual cortex as the exemplar system and revealed quantitative target-specific, layer-specific and cell-class-specific differences in its presynaptic connectomes. The retrograde connectivity indicates the presence of ventral and dorsal visual streams and further reveals topographically organized and continuously varying subnetworks mediated by different higher visual areas. The visual cortex hierarchy can be derived from intracortical feedforward and feedback pathways mediated by upper-layer and lower-layer input neurons. We also identify a new role for layer 6 neurons in mediating reciprocal interhemispheric connections. This study expands our knowledge of the visual system connectomes and demonstrates that the pipeline can be scaled up to dissect connectivity of different cell populations across the mouse brain.


Subject(s)
Connectome , Visual Cortex , Mice , Animals , Neurons/physiology , Brain/physiology , Visual Cortex/physiology , Visual Pathways
6.
Science ; 382(6667): eadf6484, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37824669

ABSTRACT

Human cortex transcriptomic studies have revealed a hierarchical organization of γ-aminobutyric acid-producing (GABAergic) neurons from subclasses to a high diversity of more granular types. Rapid GABAergic neuron viral genetic labeling plus Patch-seq (patch-clamp electrophysiology plus single-cell RNA sequencing) sampling in human brain slices was used to reliably target and analyze GABAergic neuron subclasses and individual transcriptomic types. This characterization elucidated transitions between PVALB and SST subclasses, revealed morphological heterogeneity within an abundant transcriptomic type, identified multiple spatially distinct types of the primate-specialized double bouquet cells (DBCs), and shed light on cellular differences between homologous mouse and human neocortical GABAergic neuron types. These results highlight the importance of multimodal phenotypic characterization for refinement of emerging transcriptomic cell type taxonomies and for understanding conserved and specialized cellular properties of human brain cell types.


Subject(s)
GABAergic Neurons , Interneurons , Neocortex , Animals , Humans , Mice , Electrophysiological Phenomena , GABAergic Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Interneurons/metabolism , Neocortex/cytology , Neocortex/metabolism , Patch-Clamp Techniques
7.
Neuron ; 109(9): 1449-1464.e13, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33789083

ABSTRACT

Rapid cell type identification by new genomic single-cell analysis methods has not been met with efficient experimental access to these cell types. To facilitate access to specific neural populations in mouse cortex, we collected chromatin accessibility data from individual cells and identified enhancers specific for cell subclasses and types. When cloned into recombinant adeno-associated viruses (AAVs) and delivered to the brain, these enhancers drive transgene expression in specific cortical cell subclasses. We extensively characterized several enhancer AAVs to show that they label different projection neuron subclasses as well as a homologous neuron subclass in human cortical slices. We also show how coupling enhancer viruses expressing recombinases to a newly generated transgenic mouse, Ai213, enables strong labeling of three different neuronal classes/subclasses in the brain of a single transgenic animal. This approach combines unprecedented flexibility with specificity for investigation of cell types in the mouse brain and beyond.


Subject(s)
Brain/cytology , Neurons/classification , Neurons/cytology , Single-Cell Analysis/methods , Animals , Datasets as Topic , Dependovirus , Humans , Mice , Mice, Transgenic
8.
Cell Rep ; 34(13): 108754, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33789096

ABSTRACT

Viral genetic tools that target specific brain cell types could transform basic neuroscience and targeted gene therapy. Here, we use comparative open chromatin analysis to identify thousands of human-neocortical-subclass-specific putative enhancers from across the genome to control gene expression in adeno-associated virus (AAV) vectors. The cellular specificity of reporter expression from enhancer-AAVs is established by molecular profiling after systemic AAV delivery in mouse. Over 30% of enhancer-AAVs produce specific expression in the targeted subclass, including both excitatory and inhibitory subclasses. We present a collection of Parvalbumin (PVALB) enhancer-AAVs that show highly enriched expression not only in cortical PVALB cells but also in some subcortical PVALB populations. Five vectors maintain PVALB-enriched expression in primate neocortex. These results demonstrate how genome-wide open chromatin data mining and cross-species AAV validation can be used to create the next generation of non-species-restricted viral genetic tools.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Neocortex/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Databases, Genetic , Dependovirus/genetics , Disease/genetics , Epigenesis, Genetic , Genetic Vectors/metabolism , Genome , Humans , Mice , Neurons/metabolism , Parvalbumins/metabolism , Primates , Species Specificity
9.
J Comp Neurol ; 522(9): 1989-2012, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24639291

ABSTRACT

As an anterograde neuronal tracer, recombinant adeno-associated virus (AAV) has distinct advantages over the widely used biotinylated dextran amine (BDA). However, the sensitivity and selectivity of AAV remain uncharacterized for many brain regions and species. To validate this tracing method further, AAV (serotype 1) was systematically compared with BDA as an anterograde tracer by injecting both tracers into three cortical and 15 subcortical regions in C57BL/6J mice. Identical parameters were used for our sequential iontophoretic injections, producing injections of AAV that were more robust in size and in density of neurons infected compared with those of BDA. However, these differences did not preclude further comparison between the tracers, because the pairs of injections were suitably colocalized and contained some percentage of double-labeled neurons. A qualitative analysis of projection patterns showed that the two tracers behave very similarly when injection sites are well matched. Additionally, a quantitative analysis of relative projection intensity for cases targeting primary motor cortex (MOp), primary somatosensory cortex (SSp), and caudoputamen (CP) showed strong agreement in the ranked order of projection intensities between the two tracers. A detailed analysis of the projections of two brain regions (SSp and MOp) revealed many targets that have not previously been described in the mouse or rat. Minor retrograde labeling of neurons was observed in all cases examined, for both AAV and BDA. Our results show that AAV has actions equivalent to those of BDA as an anterograde tracer and is suitable for analysis of neural circuitry throughout the mouse brain.


Subject(s)
Biotin/analogs & derivatives , Brain/anatomy & histology , Dependovirus , Dextrans , Fluorescent Dyes , Neuroanatomical Tract-Tracing Techniques , Neuronal Tract-Tracers , Animals , Cell Count , Immunohistochemistry , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence , Neural Pathways/anatomy & histology , Neurons/cytology , Photomicrography , Sensitivity and Specificity
10.
Article in English | MEDLINE | ID: mdl-25071457

ABSTRACT

Significant advances in circuit-level analyses of the brain require tools that allow for labeling, modulation of gene expression, and monitoring and manipulation of cellular activity in specific cell types and/or anatomical regions. Large-scale projects and individual laboratories have produced hundreds of gene-specific promoter-driven Cre mouse lines invaluable for enabling genetic access to subpopulations of cells in the brain. However, the potential utility of each line may not be fully realized without systematic whole brain characterization of transgene expression patterns. We established a high-throughput in situ hybridization (ISH), imaging and data processing pipeline to describe whole brain gene expression patterns in Cre driver mice. Currently, anatomical data from over 100 Cre driver lines are publicly available via the Allen Institute's Transgenic Characterization database, which can be used to assist researchers in choosing the appropriate Cre drivers for functional, molecular, or connectional studies of different regions and/or cell types in the brain.


Subject(s)
Brain/anatomy & histology , Gene Expression Regulation/physiology , Integrases/metabolism , Neurons/metabolism , Recombination, Genetic , Animals , Brain/metabolism , Gene Expression Regulation/drug effects , Integrases/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/classification , Tamoxifen/pharmacology , Trimethoprim/pharmacology
11.
J Comp Neurol ; 519(11): 2061-89, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21491433

ABSTRACT

The disrupted cortical lamination phenotype in reeler mice and subsequent identification of the Reelin signaling pathway have strongly informed models of cortical development. We describe here a marker-based phenotyping approach to reexamine the cytoarchitectural consequences of Reelin deficiency, using high-throughput histology and newly identified panels of highly specific molecular markers. The resulting cell-type-level cytoarchitectural analysis revealed novel features of abnormal patterning in the male reeler mouse not obvious with less specific markers or histology. The reeler cortex has been described as a rough laminar inversion, but the data presented here are not compatible with this model. The reeler cortex is disrupted in a more complex fashion, with some regions showing a mirror-image laminar phenotype. Major rostrocaudal and cell-type-specific differences in the laminar phenotype between cortical areas are detailed. These and similar findings in hippocampus and amygdala have implications for mechanisms of normal brain development and abnormalities in neurodevelopmental disorders.


Subject(s)
Amygdala/cytology , Cell Adhesion Molecules, Neuronal/deficiency , Extracellular Matrix Proteins/deficiency , Hippocampus/cytology , Neocortex/cytology , Nerve Tissue Proteins/deficiency , Serine Endopeptidases/deficiency , Amygdala/abnormalities , Amygdala/growth & development , Amygdala/metabolism , Animals , Biomarkers/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Hippocampus/abnormalities , Hippocampus/growth & development , Hippocampus/metabolism , In Situ Hybridization , Male , Mice , Mice, Neurologic Mutants , Neocortex/abnormalities , Neocortex/growth & development , Neocortex/metabolism , Nerve Tissue Proteins/genetics , Phenotype , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction/physiology
12.
Genome Biol ; 9(1): R23, 2008 Jan 30.
Article in English | MEDLINE | ID: mdl-18234097

ABSTRACT

With the emergence of genome-wide colorimetric in situ hybridization (ISH) data sets such as the Allen Brain Atlas, it is important to understand the relationship between this gene expression modality and those derived from more quantitative based technologies. This study introduces a novel method for standardized relative quantification of colorimetric ISH signal that enables a large-scale cross-platform expression level comparison of ISH with two publicly available microarray brain data sources.


Subject(s)
Genomics/methods , In Situ Hybridization , Oligonucleotide Array Sequence Analysis , Animals , Brain Chemistry/genetics , Colorimetry , Gene Expression Profiling , Mice , Mice, Inbred C57BL
13.
Neuron ; 60(6): 1010-21, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109908

ABSTRACT

Availability of genome-scale in situ hybridization data allows systematic analysis of genetic neuroanatomical architecture. Within the hippocampus, electrophysiology and lesion and imaging studies demonstrate functional heterogeneity along the septotemporal axis, although precise underlying circuitry and molecular substrates remain uncharacterized. Application of unbiased statistical component analyses to genome-scale hippocampal gene expression data revealed robust septotemporal molecular heterogeneity, leading to the identification of a large cohort of genes with robust regionalized hippocampal expression. Manual mapping of heterogeneous CA3 pyramidal neuron expression patterns demonstrates an unexpectedly complex molecular parcellation into a relatively coherent set of nine expression domains in the septal/temporal and proximal/distal axes with reciprocal, nonoverlapping boundaries. Unique combinatorial profiles of adhesion molecules within these domains suggest corresponding differential connectivity, which is demonstrated for CA3 projections to the lateral septum using retrograde labeling. This complex, discrete molecular architecture provides a novel paradigm for predicting functional differentiation across the full septotemporal extent of the hippocampus.


Subject(s)
Brain Mapping , Gene Expression Regulation, Developmental/physiology , Genomics , Hippocampus/anatomy & histology , Hippocampus/physiology , Animals , Animals, Newborn , Cholera Toxin/metabolism , Imaging, Three-Dimensional , In Situ Hybridization/methods , Male , Mice , Mice, Inbred C57BL , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Neural Pathways/anatomy & histology , Neural Pathways/metabolism , Principal Component Analysis , Septum Pellucidum/anatomy & histology , Septum Pellucidum/metabolism , Temporal Lobe/anatomy & histology , Temporal Lobe/metabolism
14.
Proc Natl Acad Sci U S A ; 100(8): 4903-8, 2003 Apr 15.
Article in English | MEDLINE | ID: mdl-12679517

ABSTRACT

Diverse members of the G protein-coupled receptor (GPCR) superfamily participate in a variety of physiological functions and are major targets of pharmaceutical drugs. Here we report that the repertoire of GPCRs for endogenous ligands consists of 367 receptors in humans and 392 in mice. Included here are 26 human and 83 mouse GPCRs not previously identified. A direct comparison of GPCRs in the two species reveals an unexpected level of orthology. The evolutionary preservation of these molecules argues against functional redundancy among highly related receptors. Phylogenetic analyses cluster 60% of GPCRs according to ligand preference, allowing prediction of ligand types for dozens of orphan receptors. Expression profiling of 100 GPCRs demonstrates that most are expressed in multiple tissues and that individual tissues express multiple GPCRs. Over 90% of GPCRs are expressed in the brain. Strikingly, however, the profiles of most GPCRs are unique, yielding thousands of tissue- and cell-specific receptor combinations for the modulation of physiological processes.


Subject(s)
GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Animals , Brain/metabolism , DNA, Complementary/genetics , Gene Expression , Humans , In Situ Hybridization , Ligands , Mice , Molecular Sequence Data , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL