Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ed Lein is active.

Publication


Featured researches published by Ed Lein.


Nature Neuroscience | 2010

A robust and high-throughput Cre reporting and characterization system for the whole mouse brain

Linda Madisen; Theresa A. Zwingman; Susan M. Sunkin; Seung Wook Oh; Hatim A. Zariwala; Hong Gu; Lydia Ng; Richard D. Palmiter; Michael Hawrylycz; Allan R. Jones; Ed Lein; Hongkui Zeng

The Cre/lox system is widely used in mice to achieve cell-type-specific gene expression. However, a strong and universally responding system to express genes under Cre control is still lacking. We have generated a set of Cre reporter mice with strong, ubiquitous expression of fluorescent proteins of different spectra. The robust native fluorescence of these reporters enables direct visualization of fine dendritic structures and axonal projections of the labeled neurons, which is useful in mapping neuronal circuitry, imaging and tracking specific cell populations in vivo. Using these reporters and a high-throughput in situ hybridization platform, we are systematically profiling Cre-directed gene expression throughout the mouse brain in several Cre-driver lines, including new Cre lines targeting different cell types in the cortex. Our expression data are displayed in a public online database to help researchers assess the utility of various Cre-driver lines for cell-type-specific genetic manipulation.


Nature | 2014

The complete genome sequence of a Neanderthal from the Altai Mountains

Kay Prüfer; Fernando Racimo; Nick Patterson; Flora Jay; Sriram Sankararaman; Susanna Sawyer; Anja Heinze; Gabriel Renaud; Peter H. Sudmant; Cesare de Filippo; Heng Li; Swapan Mallick; Michael Dannemann; Qiaomei Fu; Martin Kircher; Martin Kuhlwilm; Michael Lachmann; Matthias Meyer; Matthias Ongyerth; Michael Siebauer; Christoph Theunert; Arti Tandon; Priya Moorjani; Joseph K. Pickrell; James C. Mullikin; Samuel H. Vohr; Richard E. Green; Ines Hellmann; Philip L. F. Johnson; Hélène Blanché

We present a high-quality genome sequence of a Neanderthal woman from Siberia. We show that her parents were related at the level of half-siblings and that mating among close relatives was common among her recent ancestors. We also sequenced the genome of a Neanderthal from the Caucasus to low coverage. An analysis of the relationships and population history of available archaic genomes and 25 present-day human genomes shows that several gene flow events occurred among Neanderthals, Denisovans and early modern humans, possibly including gene flow into Denisovans from an unknown archaic group. Thus, interbreeding, albeit of low magnitude, occurred among many hominin groups in the Late Pleistocene. In addition, the high-quality Neanderthal genome allows us to establish a definitive list of substitutions that became fixed in modern humans after their separation from the ancestors of Neanderthals and Denisovans.


Nature | 2012

An anatomically comprehensive atlas of the adult human brain transcriptome

Michael Hawrylycz; Ed Lein; Angela L. Guillozet-Bongaarts; Elaine H. Shen; Lydia Ng; Jeremy A. Miller; Louie N. van de Lagemaat; Kimberly A. Smith; Amanda Ebbert; Zackery L. Riley; Chris Abajian; Christian F. Beckmann; Amy Bernard; Darren Bertagnolli; Andrew F. Boe; Preston M. Cartagena; M. Mallar Chakravarty; Mike Chapin; Jimmy Chong; Rachel A. Dalley; Barry Daly; Chinh Dang; Suvro Datta; Nick Dee; Tim Dolbeare; Vance Faber; David Feng; David Fowler; Jeff Goldy; Benjamin W. Gregor

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.


Nature | 2014

Transcriptional landscape of the prenatal human brain

Jeremy A. Miller; Song Lin Ding; Susan M. Sunkin; Kimberly A. Smith; Lydia Ng; Aaron Szafer; Amanda Ebbert; Zackery L. Riley; Joshua J. Royall; Kaylynn Aiona; James M. Arnold; Crissa Bennet; Darren Bertagnolli; Krissy Brouner; Stephanie Butler; Shiella Caldejon; Anita Carey; Christine Cuhaciyan; Rachel A. Dalley; Nick Dee; Tim Dolbeare; Benjamin Facer; David Feng; Tim P. Fliss; Garrett Gee; Jeff Goldy; Lindsey Gourley; Benjamin W. Gregor; Guangyu Gu; Robert Howard

The anatomical and functional architecture of the human brain is mainly determined by prenatal transcriptional processes. We describe an anatomically comprehensive atlas of the mid-gestational human brain, including de novo reference atlases, in situ hybridization, ultra-high-resolution magnetic resonance imaging (MRI) and microarray analysis on highly discrete laser-microdissected brain regions. In developing cerebral cortex, transcriptional differences are found between different proliferative and post-mitotic layers, wherein laminar signatures reflect cellular composition and developmental processes. Cytoarchitectural differences between human and mouse have molecular correlates, including species differences in gene expression in subplate, although surprisingly we find minimal differences between the inner and outer subventricular zones even though the outer zone is expanded in humans. Both germinal and post-mitotic cortical layers exhibit fronto-temporal gradients, with particular enrichment in the frontal lobe. Finally, many neurodevelopmental disorder and human-evolution-related genes show patterned expression, potentially underlying unique features of human cortical formation. These data provide a rich, freely-accessible resource for understanding human brain development.


The New England Journal of Medicine | 2014

Patches of Disorganization in the Neocortex of Children with Autism

Rich Stoner; Maggie L. Chow; Maureen P. Boyle; Susan M. Sunkin; Peter R. Mouton; Subhojit Roy; Anthony Wynshaw-Boris; Sophia A. Colamarino; Ed Lein; Eric Courchesne

BACKGROUND Autism involves early brain overgrowth and dysfunction, which is most strongly evident in the prefrontal cortex. As assessed on pathological analysis, an excess of neurons in the prefrontal cortex among children with autism signals a disturbance in prenatal development and may be concomitant with abnormal cell type and laminar development. METHODS To systematically examine neocortical architecture during the early years after the onset of autism, we used RNA in situ hybridization with a panel of layer- and cell-type-specific molecular markers to phenotype cortical microstructure. We assayed markers for neurons and glia, along with genes that have been implicated in the risk of autism, in prefrontal, temporal, and occipital neocortical tissue from postmortem samples obtained from children with autism and unaffected children between the ages of 2 and 15 years. RESULTS We observed focal patches of abnormal laminar cytoarchitecture and cortical disorganization of neurons, but not glia, in prefrontal and temporal cortical tissue from 10 of 11 children with autism and from 1 of 11 unaffected children. We observed heterogeneity between cases with respect to cell types that were most abnormal in the patches and the layers that were most affected by the pathological features. No cortical layer was uniformly spared, with the clearest signs of abnormal expression in layers 4 and 5. Three-dimensional reconstruction of layer markers confirmed the focal geometry and size of patches. CONCLUSIONS In this small, explorative study, we found focal disruption of cortical laminar architecture in the cortexes of a majority of young children with autism. Our data support a probable dysregulation of layer formation and layer-specific neuronal differentiation at prenatal developmental stages. (Funded by the Simons Foundation and others.).


Nature Neuroscience | 2011

Differential connectivity and response dynamics of excitatory and inhibitory neurons in visual cortex.

Sonja B. Hofer; Ho Ko; Bruno Pichler; Joshua T. Vogelstein; Hana Roš; Hongkui Zeng; Ed Lein; Nicholas A. Lesica; Thomas D. Mrsic-Flogel

Neuronal responses during sensory processing are influenced by both the organization of intracortical connections and the statistical features of sensory stimuli. How these intrinsic and extrinsic factors govern the activity of excitatory and inhibitory populations is unclear. Using two-photon calcium imaging in vivo and intracellular recordings in vitro, we investigated the dependencies between synaptic connectivity, feature selectivity and network activity in pyramidal cells and fast-spiking parvalbumin-expressing (PV) interneurons in mouse visual cortex. In pyramidal cell populations, patterns of neuronal correlations were largely stimulus-dependent, indicating that their responses were not strongly dominated by functionally biased recurrent connectivity. By contrast, visual stimulation only weakly modified co-activation patterns of fast-spiking PV cells, consistent with the observation that these broadly tuned interneurons received very dense and strong synaptic input from nearby pyramidal cells with diverse feature selectivities. Therefore, feedforward and recurrent network influences determine the activity of excitatory and inhibitory ensembles in fundamentally different ways.Neuronal responses during sensory processing are influenced by both the organization of intracortical connections and the statistical features of sensory stimuli. How these intrinsic and extrinsic factors govern the activity of excitatory and inhibitory populations is unclear. Using two-photon calcium imaging in vivo and intracellular recordings in vitro, we investigated the dependencies between synaptic connectivity, feature selectivity and network activity in pyramidal cells and fast-spiking parvalbumin-expressing (PV) interneurons in mouse visual cortex. In pyramidal cell populations, patterns of neuronal correlations were largely stimulus-dependent, indicating that their responses were not strongly dominated by functionally biased recurrent connectivity. By contrast, visual stimulation only weakly modified co-activation patterns of fast-spiking PV cells, consistent with the observation that these broadly tuned interneurons received very dense and strong synaptic input from nearby pyramidal cells with diverse feature selectivities. Therefore, feedforward and recurrent network influences determine the activity of excitatory and inhibitory ensembles in fundamentally different ways.


Cell | 2014

Disruptive CHD8 mutations define a subtype of autism early in development.

Raphael Bernier; Christelle Golzio; Bo Xiong; Holly A.F. Stessman; Bradley P. Coe; Osnat Penn; Kali Witherspoon; Jennifer Gerdts; Carl Baker; Anneke T. Vulto-van Silfhout; Janneke H M Schuurs-Hoeijmakers; Marco Fichera; Paolo Bosco; Serafino Buono; Antonino Alberti; Pinella Failla; Hilde Peeters; Jean Steyaert; Lisenka E.L.M. Vissers; Ludmila Francescatto; Mefford Hc; Jill A. Rosenfeld; Trygve E. Bakken; Brian J. O'Roak; Matthew Pawlus; Randall T. Moon; Jay Shendure; David G. Amaral; Ed Lein; Julia Rankin

Autism spectrum disorder (ASD) is a heterogeneous disease in which efforts to define subtypes behaviorally have met with limited success. Hypothesizing that genetically based subtype identification may prove more productive, we resequenced the ASD-associated gene CHD8 in 3,730 children with developmental delay or ASD. We identified a total of 15 independent mutations; no truncating events were identified in 8,792 controls, including 2,289 unaffected siblings. In addition to a high likelihood of an ASD diagnosis among patients bearing CHD8 mutations, characteristics enriched in this group included macrocephaly, distinct faces, and gastrointestinal complaints. chd8 disruption in zebrafish recapitulates features of the human phenotype, including increased head size as a result of expansion of the forebrain/midbrain and impairment of gastrointestinal motility due to a reduction in postmitotic enteric neurons. Our findings indicate that CHD8 disruptions define a distinct ASD subtype and reveal unexpected comorbidities between brain development and enteric innervation.


The Journal of Neuroscience | 2004

Defining a Molecular Atlas of the Hippocampus Using DNA Microarrays and High-Throughput In Situ Hybridization

Ed Lein; Xinyu Zhao; Fred H. Gage

The hippocampus consists of a series of cytoarchitecturally discrete subregions that can be distinguished from one another on the basis of morphology, connectivity, and electrophysiological properties. To understand the molecular underpinnings for these differences, DNA microarrays were used to find genes predicted to be enriched in subregions CA1, CA3, and the dentate gyrus, and >100 of these genes were subsequently analyzed using in situ hybridization to obtain cellular-level localization of their transcripts. The most striking commonality among the resulting patterns of gene expression is the extent to which cytoarchitectural boundaries within the hippocampus are respected, although the complexity of these patterns could not have been predicted on the basis of the microarray data alone. Among those genes with expression that can be characterized as “restricted” to neurons in one or more subregions of the hippocampus are a number of signal transduction molecules, transcription factors, calcium-binding proteins, and carbohydrate-modifying enzymes. These results suggest that important determinants of the unique identities of adult hippocampal neurons are differential signal transduction, regulation of gene expression, calcium homeostasis, and the maintenance of a unique extracellular milieu. Furthermore, the extremely high correlation between microarray data and in situ expression demonstrates the great utility of using DNA microarrays to genetically profile discrete brain regions.


Neuron | 2008

Genomic Anatomy of the Hippocampus

Carol L. Thompson; Sayan D. Pathak; Andreas Jeromin; Lydia Ng; Cameron Ross MacPherson; Marty T. Mortrud; Allison Cusick; Zackery L. Riley; Susan M. Sunkin; Amy Bernard; Ralph B. Puchalski; Fred H. Gage; Allan R. Jones; Vladimir B. Bajic; Michael Hawrylycz; Ed Lein

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.


Science | 2014

Convergent transcriptional specializations in the brains of humans and song-learning birds.

Andreas R. Pfenning; Erina Hara; Osceola Whitney; Miriam V. Rivas; Rui Wang; Petra L. Roulhac; Jason T. Howard; Morgan Wirthlin; Peter V. Lovell; Ganeshkumar Ganapathy; Jacquelyn Mouncastle; M. Arthur Moseley; J. Will Thompson; Erik J. Soderblom; Atsushi Iriki; Masaki Kato; M. Thomas P. Gilbert; Guojie Zhang; Trygve E. Bakken; Angie Bongaarts; Amy Bernard; Ed Lein; Claudio V. Mello; Alexander J. Hartemink; Erich D. Jarvis

INTRODUCTION Vocal learning, the ability to imitate sounds, is a trait that has undergone convergent evolution in several lineages of birds and mammals, including song-learning birds and humans. This behavior requires cortical and striatal vocal brain regions, which form unique connections in vocal-learning species. These regions have been found to have specialized gene expression within some species, but the patterns of specialization across vocal-learning bird and mammal species have not been systematically explored. Identifying molecular brain similarities across species. Brain region gene expression specializations were hierarchically organized into specialization trees of each species (blue lines), including for circuits that control learned vocalizations (highlighted green, purple, and orange regions). A set of comparative genomic algorithms found the most similarly specialized regions between songbird and human (orange lines), some of which are convergently evolved. RATIONALE The sequencing of genomes representing all major vocal-learning and vocal-nonlearning avian lineages has allowed us to develop the genomic tools to measure anatomical gene expression across species. Here, we asked whether behavioral and anatomical convergence is associated with gene expression convergence in the brains of vocal-learning birds and humans. RESULTS We developed a computational approach that discovers homologous and convergent specialized anatomical gene expression profiles. This includes generating hierarchically organized gene expression specialization trees for each species and a dynamic programming algorithm that finds the optimal alignment between species brain trees. We applied this approach to brain region gene expression databases of thousands of samples and genes that we and others generated from multiple species, including humans and song-learning birds (songbird, parrot, and hummingbird) as well as vocal-nonlearning nonhuman primates (macaque) and birds (dove and quail). Our results confirmed the recently revised understanding of the relationships between avian and mammalian brains. We further found that songbird Area X, a striatal region necessary for vocal learning, was most similar to a part of the human striatum activated during speech production. The RA (robust nucleus of the arcopallium) analog of song-learning birds, necessary for song production, was most similar to laryngeal motor cortex regions in humans that control speech production. More than 50 genes contributed to their convergent specialization and were enriched in motor control and neural connectivity functions. These patterns were not found in vocal nonlearners, but songbird RA was similar to layer 5 of primate motor cortex for another set of genes, supporting previous hypotheses about the similarity of these cell types between bird and mammal brains. CONCLUSION Our approach can accurately and quantitatively identify functionally and molecularly analogous brain regions between species separated by as much as 310 million years from a common ancestor. We were able to identify analogous brain regions for song and speech between birds and humans, and broader homologous brain regions in which these specialized song and speech regions are located, for tens to hundreds of genes. These genes now serve as candidates involved in developing and maintaining the unique connectivity and functional properties of vocal-learning brain circuits shared across species. The finding that convergent neural circuits for vocal learning are accompanied by convergent molecular changes of multiple genes in species separated by millions of years from a common ancestor indicates that brain circuits for complex traits may have limited ways in which they could have evolved from that ancestor. Song-learning birds and humans share independently evolved similarities in brain pathways for vocal learning that are essential for song and speech and are not found in most other species. Comparisons of brain transcriptomes of song-learning birds and humans relative to vocal nonlearners identified convergent gene expression specializations in specific song and speech brain regions of avian vocal learners and humans. The strongest shared profiles relate bird motor and striatal song-learning nuclei, respectively, with human laryngeal motor cortex and parts of the striatum that control speech production and learning. Most of the associated genes function in motor control and brain connectivity. Thus, convergent behavior and neural connectivity for a complex trait are associated with convergent specialized expression of multiple genes.

Collaboration


Dive into the Ed Lein's collaboration.

Top Co-Authors

Avatar

Michael Hawrylycz

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Jeremy A. Miller

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Susan M. Sunkin

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Lydia Ng

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Allan R. Jones

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Amy Bernard

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Kimberly A. Smith

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Trygve E. Bakken

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Rachel A. Dalley

Allen Institute for Brain Science

View shared research outputs
Top Co-Authors

Avatar

Darren Bertagnolli

Allen Institute for Brain Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge