101
|
Gore BB, Soden ME, Zweifel LS. Manipulating gene expression in projection-specific neuronal populations using combinatorial viral approaches. ACTA ACUST UNITED AC 2016; 65:4.35.1-20. [PMID: 25429312 DOI: 10.1002/0471142301.ns0435s65] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The mammalian brain contains tremendous structural and genetic complexity that is vital for its function. The elucidation of gene expression profiles in the brain, coupled with the development of large-scale connectivity maps and emerging viral vector-based approaches for target-selective gene manipulation, now allow for detailed dissection of gene-circuit interfaces. This protocol details how to perform combinatorial viral injections to manipulate gene expression in subsets of neurons interconnecting two brain regions. This method utilizes stereotaxic injection of a retrograde transducing CAV2-Cre virus into one brain region, combined with injection of a locally transducing Cre-dependent AAV virus into another brain region. This technique is widely applicable to the genetic dissection of neural circuitry, as it enables selective expression of candidate genes, dominant-negatives, fluorescent reporters, or genetic tools within heterogeneous populations of neurons based upon their projection targets.
Collapse
Affiliation(s)
- Bryan B Gore
- Department of Pharmacology and Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Marta E Soden
- Department of Pharmacology and Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| | - Larry S Zweifel
- Department of Pharmacology and Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA
| |
Collapse
|
102
|
Bakken TE, Miller JA, Ding SL, Sunkin SM, Smith KA, Ng L, Szafer A, Dalley RA, Royall JJ, Lemon T, Shapouri S, Aiona K, Arnold J, Bennett JL, Bertagnolli D, Bickley K, Boe A, Brouner K, Butler S, Byrnes E, Caldejon S, Carey A, Cate S, Chapin M, Chen J, Dee N, Desta T, Dolbeare TA, Dotson N, Ebbert A, Fulfs E, Gee G, Gilbert TL, Goldy J, Gourley L, Gregor B, Gu G, Hall J, Haradon Z, Haynor DR, Hejazinia N, Hoerder-Suabedissen A, Howard R, Jochim J, Kinnunen M, Kriedberg A, Kuan CL, Lau C, Lee CK, Lee F, Luong L, Mastan N, May R, Melchor J, Mosqueda N, Mott E, Ngo K, Nyhus J, Oldre A, Olson E, Parente J, Parker PD, Parry S, Pendergraft J, Potekhina L, Reding M, Riley ZL, Roberts T, Rogers B, Roll K, Rosen D, Sandman D, Sarreal M, Shapovalova N, Shi S, Sjoquist N, Sodt AJ, Townsend R, Velasquez L, Wagley U, Wakeman WB, White C, Bennett C, Wu J, Young R, Youngstrom BL, Wohnoutka P, Gibbs RA, Rogers J, Hohmann JG, Hawrylycz MJ, Hevner RF, Molnár Z, Phillips JW, Dang C, Jones AR, Amaral DG, Bernard A, Lein ES. A comprehensive transcriptional map of primate brain development. Nature 2016; 535:367-75. [PMID: 27409810 PMCID: PMC5325728 DOI: 10.1038/nature18637] [Citation(s) in RCA: 266] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 06/10/2016] [Indexed: 12/20/2022]
Abstract
The transcriptional underpinnings of brain development remain poorly understood, particularly in humans and closely related non-human primates. We describe a high-resolution transcriptional atlas of rhesus monkey (Macaca mulatta) brain development that combines dense temporal sampling of prenatal and postnatal periods with fine anatomical division of cortical and subcortical regions associated with human neuropsychiatric disease. Gene expression changes more rapidly before birth, both in progenitor cells and maturing neurons. Cortical layers and areas acquire adult-like molecular profiles surprisingly late in postnatal development. Disparate cell populations exhibit distinct developmental timing of gene expression, but also unexpected synchrony of processes underlying neural circuit construction including cell projection and adhesion. Candidate risk genes for neurodevelopmental disorders including primary microcephaly, autism spectrum disorder, intellectual disability, and schizophrenia show disease-specific spatiotemporal enrichment within developing neocortex. Human developmental expression trajectories are more similar to monkey than rodent, although approximately 9% of genes show human-specific regulation with evidence for prolonged maturation or neoteny compared to monkey.
Collapse
Affiliation(s)
- Trygve E. Bakken
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jeremy A. Miller
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Song-Lin Ding
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Susan M. Sunkin
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | | | - Lydia Ng
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Aaron Szafer
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Rachel A. Dalley
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Joshua J. Royall
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Tracy Lemon
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Sheila Shapouri
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Kaylynn Aiona
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - James Arnold
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jeffrey L. Bennett
- Department of Psychiatry and Behavioral Science, California National Primate Research Center, The M.I.N.D. Institute, University of California, Davis, Sacramento, CA 95817, USA
| | | | | | - Andrew Boe
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Krissy Brouner
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Stephanie Butler
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Emi Byrnes
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Shiella Caldejon
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Anita Carey
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Shelby Cate
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Mike Chapin
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jefferey Chen
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Tsega Desta
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Tim A. Dolbeare
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Nadia Dotson
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Amanda Ebbert
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Erich Fulfs
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Garrett Gee
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Terri L. Gilbert
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Lindsey Gourley
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Ben Gregor
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Guangyu Gu
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jon Hall
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Zeb Haradon
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - David R. Haynor
- Department of Radiology, University of Washington, Seattle, Washington 98195, USA
| | - Nika Hejazinia
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Anna Hoerder-Suabedissen
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road Oxford OX1 3QX, UK
| | - Robert Howard
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jay Jochim
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Marty Kinnunen
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Ali Kriedberg
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Chihchau L. Kuan
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Christopher Lau
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Chang-Kyu Lee
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Felix Lee
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Lon Luong
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Naveed Mastan
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Ryan May
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jose Melchor
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Nerick Mosqueda
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Erika Mott
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Kiet Ngo
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Aaron Oldre
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Eric Olson
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jody Parente
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | | | - Sheana Parry
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | | | - Lydia Potekhina
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Melissa Reding
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Zackery L. Riley
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Tyson Roberts
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Brandon Rogers
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Kate Roll
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - David Rosen
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - David Sandman
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Melaine Sarreal
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | | | - Shu Shi
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Nathan Sjoquist
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Andy J. Sodt
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Robbie Townsend
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | | | - Udi Wagley
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Wayne B. Wakeman
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Cassandra White
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Crissa Bennett
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Jennifer Wu
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Rob Young
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | | | - Paul Wohnoutka
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Richard A. Gibbs
- Human Genome Sequencing Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jeffrey Rogers
- Human Genome Sequencing Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - John G. Hohmann
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | | | - Robert F. Hevner
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, Washington 98101, USA
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road Oxford OX1 3QX, UK
| | - John W. Phillips
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Chinh Dang
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Allan R. Jones
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - David G. Amaral
- Department of Psychiatry and Behavioral Science, California National Primate Research Center, The M.I.N.D. Institute, University of California, Davis, Sacramento, CA 95817, USA
| | - Amy Bernard
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| | - Ed S. Lein
- Allen Institute for Brain Science, Seattle, Washington 98109, USA
| |
Collapse
|
103
|
Cembrowski MS, Bachman JL, Wang L, Sugino K, Shields BC, Spruston N. Spatial Gene-Expression Gradients Underlie Prominent Heterogeneity of CA1 Pyramidal Neurons. Neuron 2016; 89:351-68. [PMID: 26777276 DOI: 10.1016/j.neuron.2015.12.013] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 11/30/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023]
Abstract
Tissue and organ function has been conventionally understood in terms of the interactions among discrete and homogeneous cell types. This approach has proven difficult in neuroscience due to the marked diversity across different neuron classes, but it may be further hampered by prominent within-class variability. Here, we considered a well-defined canonical neuronal population—hippocampal CA1 pyramidal cells (CA1 PCs)—and systematically examined the extent and spatial rules of transcriptional heterogeneity. Using next-generation RNA sequencing, we identified striking variability in CA1 PCs, such that the differences within CA1 along the dorsal-ventral axis rivaled differences across distinct pyramidal neuron classes. This variability emerged from a spectrum of continuous gene-expression gradients, producing a transcriptional profile consistent with a multifarious continuum of cells. This work reveals an unexpected amount of variability within a canonical and narrowly defined neuronal population and suggests that continuous, within-class heterogeneity may be an important feature of neural circuits.
Collapse
Affiliation(s)
- Mark S Cembrowski
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, VA 20147, USA
| | | | | | | | | | | |
Collapse
|
104
|
DeFelipe J, Douglas RJ, Hill SL, Lein ES, Martin KAC, Rockland KS, Segev I, Shepherd GM, Tamás G. Comments and General Discussion on "The Anatomical Problem Posed by Brain Complexity and Size: A Potential Solution". Front Neuroanat 2016; 10:60. [PMID: 27375436 PMCID: PMC4901047 DOI: 10.3389/fnana.2016.00060] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/18/2016] [Indexed: 02/06/2023] Open
Affiliation(s)
- Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadrid, Spain; Instituto Cajal, Consejo Superior de Investigaciones CientíficasMadrid, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED)Madrid, Spain
| | - Rodney J Douglas
- Institute of Neuroinformatics, Swiss Federal Institute of Technology in Zurich (ETH) and University of Zurich (UZH) Zurich, Switzerland
| | - Sean L Hill
- Blue Brain Project, Campus Biotech Geneva, Switzerland
| | - Ed S Lein
- Human Cell Types Department, Allen Institute for Brain Science Seattle, WA, USA
| | - Kevan A C Martin
- Institute of Neuroinformatics, Swiss Federal Institute of Technology in Zurich (ETH) and University of Zurich (UZH) Zurich, Switzerland
| | - Kathleen S Rockland
- Department of Anatomy and Neurobiology, Boston University School of MedicineBoston, MA, USA; Cold Spring Harbor Laboratory, Cold Spring HarborNY, USA
| | - Idan Segev
- Departments of Neurobiology, The Hebrew University of JerusalemJerusalem, Israel; The Interdisciplinary Center for Neural Computation, The Hebrew University of JerusalemJerusalem, Israel; Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Gordon M Shepherd
- Department of Neurobiology, Yale School of Medicine New Haven, CT, USA
| | - Gábor Tamás
- MTA-SZTE Research Group for Cortical Microcircuits of the Hungarian Academy of Sciences, Department of Physiology, Anatomy and Neuroscience, University of Szeged Szeged, Hungary
| |
Collapse
|
105
|
Nagalski A, Puelles L, Dabrowski M, Wegierski T, Kuznicki J, Wisniewska MB. Molecular anatomy of the thalamic complex and the underlying transcription factors. Brain Struct Funct 2016; 221:2493-510. [PMID: 25963709 PMCID: PMC4884203 DOI: 10.1007/s00429-015-1052-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 04/27/2015] [Indexed: 01/19/2023]
Abstract
Thalamocortical loops have been implicated in the control of higher-order cognitive functions, but advances in our understanding of the molecular underpinnings of neocortical organization have not been accompanied by similar analyses in the thalamus. Using expression-based correlation maps and the manual mapping of mouse and human datasets available in the Allen Brain Atlas, we identified a few individual regions and several sets of molecularly related nuclei that partially overlap with the classic grouping that is based on topographical localization and thalamocortical connections. These new molecular divisions of the adult thalamic complex are defined by the combinatorial expression of Tcf7l2, Lef1, Gbx2, Prox1, Pou4f1, Esrrg, and Six3 transcription factor genes. Further in silico and experimental analyses provided the evidence that TCF7L2 might be a pan-thalamic specifier. These results provide substantial insights into the "molecular logic" that underlies organization of the thalamic complex.
Collapse
Affiliation(s)
- Andrzej Nagalski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, 00-927, Poland
| | - Luis Puelles
- Department of Human Anatomy, University of Murcia and IMIB, Murcia, 30071, Spain
| | - Michal Dabrowski
- Laboratory of Bioinformatics, Center of Neurobiology, Nencki Institute of Experimental Biology, Warsaw, 02-093, Poland
| | - Tomasz Wegierski
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland
| | - Jacek Kuznicki
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland
| | - Marta B Wisniewska
- Laboratory of Neurodegeneration, International Institute of Molecular and Cell Biology, Warsaw, 02-109, Poland.
- Laboratory of Molecular Neurobiology, Centre of New Technologies, University of Warsaw, Warsaw, 00-927, Poland.
| |
Collapse
|
106
|
Watakabe A. In situ hybridization analyses of claustrum-enriched genes in marmosets. J Comp Neurol 2016; 525:1442-1458. [PMID: 27098836 DOI: 10.1002/cne.24021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/15/2016] [Accepted: 04/15/2016] [Indexed: 12/13/2022]
Abstract
The claustrum/endopiriform nucleus is a unique structure that sits between the striatum and the cerebral cortex. Recent genome-wide mapping of gene expression in mice identified various genes concentrated in this structure, suggesting a requirement for a special set of genes for its function. In situ hybridization histochemistry was performed for such "claustrum-enriched" genes in the marmoset brain. In marmosets, nurr1 and netrinG2 genes exhibited highly concentrated expression in the claustrum and endopiriform nucleus, as well as in a subpopulation of layer 6 neurons across the entire cortex, consistent with their expression patterns as described in macaques. Cux2 showed enriched expression in the upper layers (layers 2-4) and the claustrum/endopiriform nucleus. GNG2 was expressed strongly in the claustrum/endopiriform nucleus, but was abundant across cortical areas in a ventral high-dorsal low gradient. Latexin was detected in the claustrum and dorsal endopiriform nucleus, but not in cortical regions. GNB4 and Tmem163 genes were both concentrated in the claustrum/endopiriform nucleus, as reported in mice, but their cortical expression in the marmoset differed from the mouse pattern. Thus, the gene set required for the claustrum appears to be broadly conserved across species, despite various differences that suggest species-specific differentiation of brain architecture. J. Comp. Neurol. 525:1442-1458, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
|
107
|
Abstract
As a species, we possess unique biological features that distinguish us from other primates. Here, we review recent efforts to identify changes in gene regulation that drove the evolution of novel human phenotypes. We discuss genotype-directed comparisons of human and nonhuman primate genomes to identify human-specific genetic changes that may encode new regulatory functions. We also review phenotype-directed approaches, which use comparisons of gene expression or regulatory function in homologous human and nonhuman primate cells and tissues to identify changes in expression levels or regulatory activity that may be due to genetic changes in humans. Together, these studies are beginning to reveal the landscape of regulatory innovation in human evolution and point to specific regulatory changes for further study. Finally, we highlight two novel strategies to model human-specific regulatory functions in vivo: primate induced pluripotent stem cells and the generation of humanized mice by genome editing.
Collapse
Affiliation(s)
- Steven K Reilly
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06510;
| | - James P Noonan
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut 06510; .,Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut 06511.,Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510
| |
Collapse
|
108
|
Abstract
The neocortex is found only in mammals, and the fossil record is silent on how this soft tissue evolved. Understanding neocortex evolution thus devolves to a search for candidate homologous neocortex traits in the extant nonmammalian amniotes. The difficulty is that homology is based on similarity, and the six-layered neocortex structure could hardly be more dissimilar in appearance from the nuclear organization that is so conspicuous in the dorsal telencephalon of birds and other reptiles. Recent molecular data have, however, provided new support for one prominent hypothesis, based on neuronal circuits, that proposes the principal neocortical input and output cell types are a conserved feature of amniote dorsal telencephalon. Many puzzles remain, the greatest being understanding the selective pressures and molecular mechanisms that underlie such tremendous morphological variation in telencephalon structure.
Collapse
Affiliation(s)
- Jennifer Dugas-Ford
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, Illinois 60637;
| | | |
Collapse
|
109
|
El-Shamayleh Y, Ni AM, Horwitz GD. Strategies for targeting primate neural circuits with viral vectors. J Neurophysiol 2016; 116:122-34. [PMID: 27052579 PMCID: PMC4961743 DOI: 10.1152/jn.00087.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/05/2016] [Indexed: 11/22/2022] Open
Abstract
Understanding how the brain works requires understanding how different types of neurons contribute to circuit function and organism behavior. Progress on this front has been accelerated by optogenetics and chemogenetics, which provide an unprecedented level of control over distinct neuronal types in small animals. In primates, however, targeting specific types of neurons with these tools remains challenging. In this review, we discuss existing and emerging strategies for directing genetic manipulations to targeted neurons in the adult primate central nervous system. We review the literature on viral vectors for gene delivery to neurons, focusing on adeno-associated viral vectors and lentiviral vectors, their tropism for different cell types, and prospects for new variants with improved efficacy and selectivity. We discuss two projection targeting approaches for probing neural circuits: anterograde projection targeting and retrograde transport of viral vectors. We conclude with an analysis of cell type-specific promoters and other nucleotide sequences that can be used in viral vectors to target neuronal types at the transcriptional level.
Collapse
Affiliation(s)
- Yasmine El-Shamayleh
- Department of Physiology and Biophysics and Washington National Primate Research Center, University of Washington, Seattle, Washington; and
| | - Amy M Ni
- Department of Neuroscience and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gregory D Horwitz
- Department of Physiology and Biophysics and Washington National Primate Research Center, University of Washington, Seattle, Washington; and
| |
Collapse
|
110
|
Miller KD. Canonical computations of cerebral cortex. Curr Opin Neurobiol 2016; 37:75-84. [PMID: 26868041 DOI: 10.1016/j.conb.2016.01.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 12/23/2022]
Abstract
The idea that there is a fundamental cortical circuit that performs canonical computations remains compelling though far from proven. Here we review evidence for two canonical operations within sensory cortical areas: a feedforward computation of selectivity; and a recurrent computation of gain in which, given sufficiently strong external input, perhaps from multiple sources, intracortical input largely, but not completely, cancels this external input. This operation leads to many characteristic cortical nonlinearities in integrating multiple stimuli. The cortical computation must combine such local processing with hierarchical processing across areas. We point to important changes in moving from sensory cortex to motor and frontal cortex and the possibility of substantial differences between cortex in rodents vs. species with columnar organization of selectivity.
Collapse
Affiliation(s)
- Kenneth D Miller
- Center for Theoretical Neuroscience, Department of Neuroscience, Swartz Program in Theoretical Neuroscience, Kavli Institute for Brain Science, College of Physicians and Surgeons, Columbia University, New York, NY 10032-2695, United States.
| |
Collapse
|
111
|
Krienen FM, Yeo BTT, Ge T, Buckner RL, Sherwood CC. Transcriptional profiles of supragranular-enriched genes associate with corticocortical network architecture in the human brain. Proc Natl Acad Sci U S A 2016; 113:E469-78. [PMID: 26739559 PMCID: PMC4739529 DOI: 10.1073/pnas.1510903113] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The human brain is patterned with disproportionately large, distributed cerebral networks that connect multiple association zones in the frontal, temporal, and parietal lobes. The expansion of the cortical surface, along with the emergence of long-range connectivity networks, may be reflected in changes to the underlying molecular architecture. Using the Allen Institute's human brain transcriptional atlas, we demonstrate that genes particularly enriched in supragranular layers of the human cerebral cortex relative to mouse distinguish major cortical classes. The topography of transcriptional expression reflects large-scale brain network organization consistent with estimates from functional connectivity MRI and anatomical tracing in nonhuman primates. Microarray expression data for genes preferentially expressed in human upper layers (II/III), but enriched only in lower layers (V/VI) of mouse, were cross-correlated to identify molecular profiles across the cerebral cortex of postmortem human brains (n = 6). Unimodal sensory and motor zones have similar molecular profiles, despite being distributed across the cortical mantle. Sensory/motor profiles were anticorrelated with paralimbic and certain distributed association network profiles. Tests of alternative gene sets did not consistently distinguish sensory and motor regions from paralimbic and association regions: (i) genes enriched in supragranular layers in both humans and mice, (ii) genes cortically enriched in humans relative to nonhuman primates, (iii) genes related to connectivity in rodents, (iv) genes associated with human and mouse connectivity, and (v) 1,454 gene sets curated from known gene ontologies. Molecular innovations of upper cortical layers may be an important component in the evolution of long-range corticocortical projections.
Collapse
Affiliation(s)
- Fenna M Krienen
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology and Institute for Neuroscience, The George Washington University, Washington, DC 20052;
| | - B T Thomas Yeo
- Department of Electrical and Computer Engineering, Clinical Imaging Research Centre, Singapore Institute for Neurotechnology & Memory Networks Program, National University of Singapore, Singapore 117583; Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129
| | - Tian Ge
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129; Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114
| | - Randy L Buckner
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA 02129; Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114; Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA 02138
| | - Chet C Sherwood
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology and Institute for Neuroscience, The George Washington University, Washington, DC 20052
| |
Collapse
|
112
|
Frackowiak R, Markram H. The future of human cerebral cartography: a novel approach. Philos Trans R Soc Lond B Biol Sci 2015; 370:rstb.2014.0171. [PMID: 25823868 PMCID: PMC4387512 DOI: 10.1098/rstb.2014.0171] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cerebral cartography can be understood in a limited, static, neuroanatomical sense. Temporal information from electrical recordings contributes information on regional interactions adding a functional dimension. Selective tagging and imaging of molecules adds biochemical contributions. Cartographic detail can also be correlated with normal or abnormal psychological or behavioural data. Modern cerebral cartography is assimilating all these elements. Cartographers continue to collect ever more precise data in the hope that general principles of organization will emerge. However, even detailed cartographic data cannot generate knowledge without a multi-scale framework making it possible to relate individual observations and discoveries. We propose that, in the next quarter century, advances in cartography will result in progressively more accurate drafts of a data-led, multi-scale model of human brain structure and function. These blueprints will result from analysis of large volumes of neuroscientific and clinical data, by a process of reconstruction, modelling and simulation. This strategy will capitalize on remarkable recent developments in informatics and computer science and on the existence of much existing, addressable data and prior, though fragmented, knowledge. The models will instantiate principles that govern how the brain is organized at different levels and how different spatio-temporal scales relate to each other in an organ-centred context.
Collapse
Affiliation(s)
- Richard Frackowiak
- The Human Brain Project, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne 1011, Switzerland
| | - Henry Markram
- The Human Brain Project, Ecole Polytechnique Fedérale de Lausanne, Lausanne 1015, Switzerland
| |
Collapse
|
113
|
Silver DL. Genomic divergence and brain evolution: How regulatory DNA influences development of the cerebral cortex. Bioessays 2015; 38:162-71. [PMID: 26642006 DOI: 10.1002/bies.201500108] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The cerebral cortex controls our most distinguishing higher cognitive functions. Human-specific gene expression differences are abundant in the cerebral cortex, yet we have only begun to understand how these variations impact brain function. This review discusses the current evidence linking non-coding regulatory DNA changes, including enhancers, with neocortical evolution. Functional interrogation using animal models reveals converging roles for our genome in key aspects of cortical development including progenitor cell cycle and neuronal signaling. New technologies, including iPS cells and organoids, offer potential alternatives to modeling evolutionary modifications in a relevant species context. Several diseases rooted in the cerebral cortex uniquely manifest in humans compared to other primates, thus highlighting the importance of understanding human brain differences. Future studies of regulatory loci, including those implicated in disease, will collectively help elucidate key cellular and genetic mechanisms underlying our distinguishing cognitive traits.
Collapse
Affiliation(s)
- Debra L Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA.,Department of Cell Biology, Duke University Medical Center, Durham, NC, USA.,Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.,Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
114
|
Hawrylycz M, Miller JA, Menon V, Feng D, Dolbeare T, Guillozet-Bongaarts AL, Jegga AG, Aronow BJ, Lee CK, Bernard A, Glasser MF, Dierker DL, Menche J, Szafer A, Collman F, Grange P, Berman KA, Mihalas S, Yao Z, Stewart L, Barabási AL, Schulkin J, Phillips J, Ng L, Dang C, Haynor DR, Jones A, Van Essen DC, Koch C, Lein E. Canonical genetic signatures of the adult human brain. Nat Neurosci 2015; 18:1832-44. [PMID: 26571460 PMCID: PMC4700510 DOI: 10.1038/nn.4171] [Citation(s) in RCA: 442] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 10/16/2015] [Indexed: 11/09/2022]
Abstract
The structure and function of the human brain are highly stereotyped, implying a conserved molecular program responsible for its development, cellular structure and function. We applied a correlation-based metric called differential stability to assess reproducibility of gene expression patterning across 132 structures in six individual brains, revealing mesoscale genetic organization. The genes with the highest differential stability are highly biologically relevant, with enrichment for brain-related annotations, disease associations, drug targets and literature citations. Using genes with high differential stability, we identified 32 anatomically diverse and reproducible gene expression signatures, which represent distinct cell types, intracellular components and/or associations with neurodevelopmental and neurodegenerative disorders. Genes in neuron-associated compared to non-neuronal networks showed higher preservation between human and mouse; however, many diversely patterned genes displayed marked shifts in regulation between species. Finally, highly consistent transcriptional architecture in neocortex is correlated with resting state functional connectivity, suggesting a link between conserved gene expression and functionally relevant circuitry.
Collapse
Affiliation(s)
| | - Jeremy A Miller
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Vilas Menon
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - David Feng
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Tim Dolbeare
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | | | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital and Medical Center, Cincinnati, Ohio, USA
| | - Bruce J Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital and Medical Center, Cincinnati, Ohio, USA
| | - Chang-Kyu Lee
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Amy Bernard
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Matthew F Glasser
- Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri, USA
| | - Donna L Dierker
- Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri, USA
| | - Jörg Menche
- Center for Complex Networks Research, Northeastern University, Boston, Massachusetts, USA.,Department of Physics, Northeastern University, Boston, Massachusetts, USA.,Center for Network Science, Central European University, Budapest, Hungary
| | - Aaron Szafer
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Forrest Collman
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Pascal Grange
- Department of Mathematical Sciences, Xi'an Jiaotong-Liverpool University, Jiangsu, China
| | - Kenneth A Berman
- Department of Electrical Engineering and Computing Systems, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stefan Mihalas
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Zizhen Yao
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Lance Stewart
- Institute for Protein Design, University of Washington, Seattle, Washington, USA
| | - Albert-László Barabási
- Center for Complex Networks Research, Northeastern University, Boston, Massachusetts, USA.,Department of Physics, Northeastern University, Boston, Massachusetts, USA.,Center for Network Science, Central European University, Budapest, Hungary.,Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jay Schulkin
- Department of Neuroscience, Georgetown University, Washington, DC, USA
| | - John Phillips
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Lydia Ng
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Chinh Dang
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - David R Haynor
- Department of Radiology, The University of Washington, Seattle, Washington, USA
| | - Allan Jones
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - David C Van Essen
- Department of Anatomy and Neurobiology, Washington University, St. Louis, Missouri, USA
| | - Christof Koch
- The Allen Institute for Brain Science, Seattle, Washington, USA
| | - Ed Lein
- The Allen Institute for Brain Science, Seattle, Washington, USA
| |
Collapse
|
115
|
Sanders SJ. First glimpses of the neurobiology of autism spectrum disorder. Curr Opin Genet Dev 2015; 33:80-92. [PMID: 26547130 DOI: 10.1016/j.gde.2015.10.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/05/2015] [Accepted: 10/07/2015] [Indexed: 12/22/2022]
Abstract
Rapid progress in identifying the genes underlying autism spectrum disorder (ASD) has provided the substrate for a first wave of analyses into the underlying neurobiology. This review describes the consensus across these diverse analyses, highlighting two distinct sets of genes: 1) Genes that regulate chromatin and transcription, especially in cortical projection neurons and striatal medium spiny neurons during mid-fetal development; and 2) Genes involved in synapse development and function, especially during infancy and early childhood, and differentially expressed in the post mortem ASD brain. Both gene sets are also regulatory targets of the ASD genes CHD8 and FMRP. It remains to be seen whether these represent two independent paths to the ASD phenotype or two components of a common path.
Collapse
Affiliation(s)
- Stephan J Sanders
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
116
|
Thompson BL, Levitt P. Complete or partial reduction of the Met receptor tyrosine kinase in distinct circuits differentially impacts mouse behavior. J Neurodev Disord 2015; 7:35. [PMID: 26523156 PMCID: PMC4628780 DOI: 10.1186/s11689-015-9131-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/20/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Our laboratory discovered that the gene encoding the receptor tyrosine kinase, MET, contributes to autism risk. Expression of MET is reduced in human postmortem temporal lobe in autism and Rett Syndrome. Subsequent studies revealed a role for MET in human and mouse functional and structural cortical connectivity. To further understand the contribution of Met to brain development and its impact on behavior, we generated two conditional mouse lines in which Met is deleted from select populations of central nervous system neurons. Mice were then tested to determine the consequences of disrupting Met expression. METHODS Mating of Emx1 (cre) and Met (fx/fx) mice eliminates receptor signaling from all cells arising from the dorsal pallium. Met (fx/fx) and Nestin (cre) crosses result in receptor signaling elimination from all neural cells. Behavioral tests were performed to assess cognitive, emotional, and social impairments that are observed in multiple neurodevelopmental disorders and that are in part subserved by circuits that express Met. RESULTS Met (fx/fx) /Emx1 (cre) null mice displayed significant hypoactivity in the activity chamber and in the T-maze despite superior performance on the rotarod. Additionally, these animals showed a deficit in spontaneous alternation. Surprisingly, Met (fx/fx; fx/+) /Nestin (cre) null and heterozygous mice exhibited deficits in contextual fear conditioning, and Met (fx/+) /Nestin (cre) heterozygous mice spent less time in the closed arms of the elevated plus maze. CONCLUSIONS These data suggest a complex contribution of Met in the development of circuits mediating social, emotional, and cognitive behavior. The impact of disrupting developmental Met expression is dependent upon circuit-specific deletion patterns and levels of receptor activity.
Collapse
Affiliation(s)
- Barbara L Thompson
- Chan Division of Occupational Science and Occupational Therapy, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90089 USA ; Institute for the Developing Mind, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027 USA ; Department of Pediatrics, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027 USA
| | - Pat Levitt
- Institute for the Developing Mind, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027 USA ; Department of Pediatrics, Children's Hospital of Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90027 USA
| |
Collapse
|
117
|
In Vivo Two-Photon Imaging of Dendritic Spines in Marmoset Neocortex. eNeuro 2015; 2:eN-MNT-0019-15. [PMID: 26465000 PMCID: PMC4596018 DOI: 10.1523/eneuro.0019-15.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 07/03/2015] [Accepted: 07/27/2015] [Indexed: 11/21/2022] Open
Abstract
Two-photon microscopy in combination with a technique involving the artificial expression of fluorescent protein has enabled the direct observation of dendritic spines in living brains. However, the application of this method to primate brains has been hindered by the lack of appropriate labeling techniques for visualizing dendritic spines. Here, we developed an adeno-associated virus vector-based fluorescent protein expression system for visualizing dendritic spines in vivo in the marmoset neocortex. For the clear visualization of each spine, the expression of reporter fluorescent protein should be both sparse and strong. To fulfill these requirements, we amplified fluorescent signals using the tetracycline transactivator (tTA)–tetracycline-responsive element system and by titrating down the amount of Thy1S promoter-driven tTA for sparse expression. By this method, we were able to visualize dendritic spines in the marmoset cortex by two-photon microscopy in vivo and analyze the turnover of spines in the prefrontal cortex. Our results demonstrated that short spines in the marmoset cortex tend to change more frequently than long spines. The comparison of in vivo samples with fixed samples showed that we did not detect all existing spines by our method. Although we found glial cell proliferation, the damage of tissues caused by window construction was relatively small, judging from the comparison of spine length between samples with or without window construction. Our new labeling technique for two-photon imaging to visualize in vivo dendritic spines of the marmoset neocortex can be applicable to examining circuit reorganization and synaptic plasticity in primates.
Collapse
|
118
|
Hackett TA, Guo Y, Clause A, Hackett NJ, Garbett K, Zhang P, Polley DB, Mirnics K. Transcriptional maturation of the mouse auditory forebrain. BMC Genomics 2015; 16:606. [PMID: 26271746 PMCID: PMC4536593 DOI: 10.1186/s12864-015-1709-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/01/2015] [Indexed: 02/07/2023] Open
Abstract
Background The maturation of the brain involves the coordinated expression of thousands of genes, proteins and regulatory elements over time. In sensory pathways, gene expression profiles are modified by age and sensory experience in a manner that differs between brain regions and cell types. In the auditory system of altricial animals, neuronal activity increases markedly after the opening of the ear canals, initiating events that culminate in the maturation of auditory circuitry in the brain. This window provides a unique opportunity to study how gene expression patterns are modified by the onset of sensory experience through maturity. As a tool for capturing these features, next-generation sequencing of total RNA (RNAseq) has tremendous utility, because the entire transcriptome can be screened to index expression of any gene. To date, whole transcriptome profiles have not been generated for any central auditory structure in any species at any age. In the present study, RNAseq was used to profile two regions of the mouse auditory forebrain (A1, primary auditory cortex; MG, medial geniculate) at key stages of postnatal development (P7, P14, P21, adult) before and after the onset of hearing (~P12). Hierarchical clustering, differential expression, and functional geneset enrichment analyses (GSEA) were used to profile the expression patterns of all genes. Selected genesets related to neurotransmission, developmental plasticity, critical periods and brain structure were highlighted. An accessible repository of the entire dataset was also constructed that permits extraction and screening of all data from the global through single-gene levels. To our knowledge, this is the first whole transcriptome sequencing study of the forebrain of any mammalian sensory system. Although the data are most relevant for the auditory system, they are generally applicable to forebrain structures in the visual and somatosensory systems, as well. Results The main findings were: (1) Global gene expression patterns were tightly clustered by postnatal age and brain region; (2) comparing A1 and MG, the total numbers of differentially expressed genes were comparable from P7 to P21, then dropped to nearly half by adulthood; (3) comparing successive age groups, the greatest numbers of differentially expressed genes were found between P7 and P14 in both regions, followed by a steady decline in numbers with age; (4) maturational trajectories in expression levels varied at the single gene level (increasing, decreasing, static, other); (5) between regions, the profiles of single genes were often asymmetric; (6) GSEA revealed that genesets related to neural activity and plasticity were typically upregulated from P7 to adult, while those related to structure tended to be downregulated; (7) GSEA and pathways analysis of selected functional networks were not predictive of expression patterns in the auditory forebrain for all genes, reflecting regional specificity at the single gene level. Conclusions Gene expression in the auditory forebrain during postnatal development is in constant flux and becomes increasingly stable with age. Maturational changes are evident at the global through single gene levels. Transcriptome profiles in A1 and MG are distinct at all ages, and differ from other brain regions. The database generated by this study provides a rich foundation for the identification of novel developmental biomarkers, functional gene pathways, and targeted studies of postnatal maturation in the auditory forebrain. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1709-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Troy A Hackett
- Department of Hearing and Speech Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37232, USA.
| | - Yan Guo
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA.
| | - Amanda Clause
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Department of Otology and Laryngology, Harvard Medical School, Boston, MA, USA.
| | | | | | - Pan Zhang
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA.
| | - Daniel B Polley
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Department of Otology and Laryngology, Harvard Medical School, Boston, MA, USA.
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA. .,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, 37235, USA. .,Department of Psychiatry, University of Szeged, 6725, Szeged, Hungary. .,Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
119
|
Bauernfeind AL, Reyzer ML, Caprioli RM, Ely JJ, Babbitt CC, Wray GA, Hof PR, Sherwood CC. High spatial resolution proteomic comparison of the brain in humans and chimpanzees. J Comp Neurol 2015; 523:2043-61. [PMID: 25779868 DOI: 10.1002/cne.23777] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/03/2015] [Accepted: 03/11/2015] [Indexed: 12/30/2022]
Abstract
We performed high-throughput mass spectrometry at high spatial resolution from individual regions (anterior cingulate and primary motor, somatosensory, and visual cortices) and layers of the neocortex (layers III, IV, and V) and cerebellum (granule cell layer), as well as the caudate nucleus in humans and chimpanzees. A total of 39 mass spectrometry peaks were matched with probable protein identifications in both species, allowing for comparison in expression. We explored how the pattern of protein expression varies across regions and cortical layers to provide insights into the differences in molecular phenotype of these neural structures between species. The expression of proteins differed principally in a region- and layer-specific pattern, with more subtle differences between species. Specifically, human and chimpanzee brains were similar in their distribution of proteins related to the regulation of transcription and enzyme activity but differed in their expression of proteins supporting aerobic metabolism. Whereas most work assessing molecular expression differences in the brains of primates has been performed on gene transcripts, this dataset extends current understanding of the differential molecular expression that may underlie human cognitive specializations.
Collapse
Affiliation(s)
- Amy L Bauernfeind
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, 63110.,Department of Anthropology, Washington University in St. Louis, St. Louis, Missouri, 63130.,Department of Anthropology, The George Washington University, Washington, DC, 20052
| | - Michelle L Reyzer
- Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Department of Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee, 37232
| | - Richard M Caprioli
- Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, Tennessee, 37232.,Department of Biochemistry, Vanderbilt University Medical Center, Nashville, Tennessee, 37232
| | - John J Ely
- MAEBIOS-TM, Alamogordo, New Mexico, 88310
| | - Courtney C Babbitt
- Department of Biology, University of Massachusetts Amherst, Amherst, Massachusetts 01003
| | - Gregory A Wray
- Institute for Genome Sciences & Policy, Duke University, Durham, North Carolina, 27708.,Department of Biology, Duke University, Durham, North Carolina, 27708.,Department of Evolutionary Anthropology, Duke University, Durham, North Carolina, 27708
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029.,New York Consortium in Evolutionary Primatology, New York, New York
| | - Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC, 20052
| |
Collapse
|
120
|
Bauernfeind AL, Soderblom EJ, Turner ME, Moseley MA, Ely JJ, Hof PR, Sherwood CC, Wray GA, Babbitt CC. Evolutionary Divergence of Gene and Protein Expression in the Brains of Humans and Chimpanzees. Genome Biol Evol 2015; 7:2276-88. [PMID: 26163674 PMCID: PMC4558850 DOI: 10.1093/gbe/evv132] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although transcriptomic profiling has become the standard approach for exploring molecular differences in the primate brain, very little is known about how the expression levels of gene transcripts relate to downstream protein abundance. Moreover, it is unknown whether the relationship changes depending on the brain region or species under investigation. We performed high-throughput transcriptomic (RNA-Seq) and proteomic (liquid chromatography coupled with tandem mass spectrometry) analyses on two regions of the human and chimpanzee brain: The anterior cingulate cortex and caudate nucleus. In both brain regions, we found a lower correlation between mRNA and protein expression levels in humans and chimpanzees than has been reported for other tissues and cell types, suggesting that the brain may engage extensive tissue-specific regulation affecting protein abundance. In both species, only a few categories of biological function exhibited strong correlations between mRNA and protein expression levels. These categories included oxidative metabolism and protein synthesis and modification, indicating that the expression levels of mRNA transcripts supporting these biological functions are more predictive of protein expression compared with other functional categories. More generally, however, the two measures of molecular expression provided strikingly divergent perspectives into differential expression between human and chimpanzee brains: mRNA comparisons revealed significant differences in neuronal communication, ion transport, and regulatory processes, whereas protein comparisons indicated differences in perception and cognition, metabolic processes, and organization of the cytoskeleton. Our results highlight the importance of examining protein expression in evolutionary analyses and call for a more thorough understanding of tissue-specific protein expression levels.
Collapse
Affiliation(s)
- Amy L Bauernfeind
- Department of Anatomy and Neurobiology, Washington University Medical School Department of Anthropology, Washington University in St. Louis Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University
| | - Erik J Soderblom
- Proteomics and Metabolomics Shared Resource, Duke University School of Medicine Center for Genomic and Computational Biology, Duke University
| | - Meredith E Turner
- Proteomics and Metabolomics Shared Resource, Duke University School of Medicine Center for Genomic and Computational Biology, Duke University
| | - M Arthur Moseley
- Proteomics and Metabolomics Shared Resource, Duke University School of Medicine Center for Genomic and Computational Biology, Duke University
| | | | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York New York Consortium in Evolutionary Primatology, New York, New York
| | - Chet C Sherwood
- Department of Anthropology and Center for the Advanced Study of Human Paleobiology, The George Washington University
| | - Gregory A Wray
- Center for Genomic and Computational Biology, Duke University Department of Biology, Duke University Department of Evolutionary Anthropology, Duke University
| | | |
Collapse
|
121
|
Abstract
Three decades ago, Rockel et al. proposed that neuronal surface densities (number of neurons under a square millimeter of surface) of primary visual cortices (V1s) in primates is 2.5 times higher than the neuronal density of V1s in nonprimates or many other cortical regions in primates and nonprimates. This claim has remained controversial and much debated. We replicated the study of Rockel et al. with attention to modern stereological precepts and show that indeed primate V1 is 2.5 times denser (number of neurons per square millimeter) than many other cortical regions and nonprimate V1s; we also show that V2 is 1.7 times as dense. As primate V1s are denser, they have more neurons and thus more pinwheels than similar-sized nonprimate V1s, which explains why primates have better visual acuity.
Collapse
|
122
|
Bakken TE, Miller JA, Luo R, Bernard A, Bennett JL, Lee CK, Bertagnolli D, Parikshak NN, Smith KA, Sunkin SM, Amaral DG, Geschwind DH, Lein ES. Spatiotemporal dynamics of the postnatal developing primate brain transcriptome. Hum Mol Genet 2015; 24:4327-39. [PMID: 25954031 PMCID: PMC4492396 DOI: 10.1093/hmg/ddv166] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/05/2015] [Indexed: 01/06/2023] Open
Abstract
Developmental changes in the temporal and spatial regulation of gene expression drive the emergence of normal mature brain function, while disruptions in these processes underlie many neurodevelopmental abnormalities. To solidify our foundational knowledge of such changes in a primate brain with an extended period of postnatal maturation like in human, we investigated the whole-genome transcriptional profiles of rhesus monkey brains from birth to adulthood. We found that gene expression dynamics are largest from birth through infancy, after which gene expression profiles transition to a relatively stable state by young adulthood. Biological pathway enrichment analysis revealed that genes more highly expressed at birth are associated with cell adhesion and neuron differentiation, while genes more highly expressed in juveniles and adults are associated with cell death. Neocortex showed significantly greater differential expression over time than subcortical structures, and this trend likely reflects the protracted postnatal development of the cortex. Using network analysis, we identified 27 co-expression modules containing genes with highly correlated expression patterns that are associated with specific brain regions, ages or both. In particular, one module with high expression in neonatal cortex and striatum that decreases during infancy and juvenile development was significantly enriched for autism spectrum disorder (ASD)-related genes. This network was enriched for genes associated with axon guidance and interneuron differentiation, consistent with a disruption in the formation of functional cortical circuitry in ASD.
Collapse
Affiliation(s)
| | | | - Rui Luo
- Human Genetics Program, Department of Neurology and Semel Institute, David Geffen School of Medicine, UC, Los Angeles, Los Angeles, CA, USA and
| | - Amy Bernard
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeffrey L Bennett
- Department of Psychiatry and Behavioral Science and M.I.N.D. Institute, UC Davis, Sacramento, CA, USA
| | | | | | - Neelroop N Parikshak
- Human Genetics Program, Department of Neurology and Semel Institute, David Geffen School of Medicine, UC, Los Angeles, Los Angeles, CA, USA and
| | | | | | - David G Amaral
- Department of Psychiatry and Behavioral Science and M.I.N.D. Institute, UC Davis, Sacramento, CA, USA
| | - Daniel H Geschwind
- Human Genetics Program, Department of Neurology and Semel Institute, David Geffen School of Medicine, UC, Los Angeles, Los Angeles, CA, USA and
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA, USA,
| |
Collapse
|
123
|
Abstract
The development and function of our brain are governed by a genetic blueprint, which reflects dynamic changes over the history of evolution. Recent progress in genetics and genomics, facilitated by next-generation sequencing and single-cell sorting, has identified numerous genomic loci that are associated with a neuroanatomical or neurobehavioral phenotype. Here, we review some of the genetic changes in both protein-coding and noncoding regions that affect brain development and evolution, as well as recent progress in brain transcriptomics. Understanding these genetic changes may provide novel insights into neurological and neuropsychiatric disorders, such as autism and schizophrenia.
Collapse
Affiliation(s)
- Byoung-Il Bae
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Boston, MA 02115, USA; and Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Divya Jayaraman
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Boston, MA 02115, USA; and Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease, and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Boston, MA 02115, USA; and Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
124
|
Telese F, Ma Q, Perez PM, Notani D, Oh S, Li W, Comoletti D, Ohgi KA, Taylor H, Rosenfeld MG. LRP8-Reelin-Regulated Neuronal Enhancer Signature Underlying Learning and Memory Formation. Neuron 2015; 86:696-710. [PMID: 25892301 DOI: 10.1016/j.neuron.2015.03.033] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 02/11/2015] [Accepted: 03/13/2015] [Indexed: 11/16/2022]
Abstract
One of the exceptional properties of the brain is its ability to acquire new knowledge through learning and to store that information through memory. The epigenetic mechanisms linking changes in neuronal transcriptional programs to behavioral plasticity remain largely unknown. Here, we identify the epigenetic signature of the neuronal enhancers required for transcriptional regulation of synaptic plasticity genes during memory formation, linking this to Reelin signaling. The binding of Reelin to its receptor, LRP8, triggers activation of this cohort of LRP8-Reelin-regulated neuronal (LRN) enhancers that serve as the ultimate convergence point of a novel synapse-to-nucleus pathway. Reelin simultaneously regulates NMDA-receptor transmission, which reciprocally permits the required γ-secretase-dependent cleavage of LRP8, revealing an unprecedented role for its intracellular domain in the regulation of synaptically generated signals. These results uncover an in vivo enhancer code serving as a critical molecular component of cognition and relevant to psychiatric disorders linked to defects in Reelin signaling.
Collapse
Affiliation(s)
- Francesca Telese
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Qi Ma
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Bioinformatis and System Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Patricia Montilla Perez
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dimple Notani
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Soohwan Oh
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Biology Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Wenbo Li
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Davide Comoletti
- Child Health Institute of New Jersey and Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Kenneth A Ohgi
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Havilah Taylor
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
125
|
Wilken MS, Brzezinski JA, La Torre A, Siebenthall K, Thurman R, Sabo P, Sandstrom RS, Vierstra J, Canfield TK, Hansen RS, Bender MA, Stamatoyannopoulos J, Reh TA. DNase I hypersensitivity analysis of the mouse brain and retina identifies region-specific regulatory elements. Epigenetics Chromatin 2015; 8:8. [PMID: 25972927 PMCID: PMC4429822 DOI: 10.1186/1756-8935-8-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/27/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The brain, spinal cord, and neural retina comprise the central nervous system (CNS) of vertebrates. Understanding the regulatory mechanisms that underlie the enormous cell-type diversity of the CNS is a significant challenge. Whole-genome mapping of DNase I-hypersensitive sites (DHSs) has been used to identify cis-regulatory elements in many tissues. We have applied this approach to the mouse CNS, including developing and mature neural retina, whole brain, and two well-characterized brain regions, the cerebellum and the cerebral cortex. RESULTS For the various regions and developmental stages of the CNS that we analyzed, there were approximately the same number of DHSs; however, there were many DHSs unique to each CNS region and developmental stage. Many of the DHSs are likely to mark enhancers that are specific to the specific CNS region and developmental stage. We validated the DNase I mapping approach for identification of CNS enhancers using the existing VISTA Browser database and with in vivo and in vitro electroporation of the retina. Analysis of transcription factor consensus sites within the DHSs shows distinct region-specific profiles of transcriptional regulators particular to each region. Clustering developmentally dynamic DHSs in the retina revealed enrichment of developmental stage-specific transcriptional regulators. Additionally, we found reporter gene activity in the retina driven from several previously uncharacterized regulatory elements surrounding the neurodevelopmental gene Otx2. Identification of DHSs shared between mouse and human showed region-specific differences in the evolution of cis-regulatory elements. CONCLUSIONS Overall, our results demonstrate the potential of genome-wide DNase I mapping to cis-regulatory questions regarding the regional diversity within the CNS. These data represent an extensive catalogue of potential cis-regulatory elements within the CNS that display region and temporal specificity, as well as a set of DHSs common to CNS tissues. Further examination of evolutionary conservation of DHSs between CNS regions and different species may reveal important cis-regulatory elements in the evolution of the mammalian CNS.
Collapse
Affiliation(s)
- Matthew S Wilken
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Box 357420, Seattle, WA 98195 USA ; Molecular and Cellular Biology Program, University of Washington, MCB Program Office, T-466 Health Sciences Building, Box 357275, Seattle, WA 98195 USA
| | - Joseph A Brzezinski
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Box 357420, Seattle, WA 98195 USA ; Department of Ophthalmology, University of Colorado School of Medicine, 1675 Aurora Court, Aurora, CO 80045 USA
| | - Anna La Torre
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Box 357420, Seattle, WA 98195 USA
| | - Kyle Siebenthall
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - Robert Thurman
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - Peter Sabo
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - Richard S Sandstrom
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - Jeff Vierstra
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - Theresa K Canfield
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - R Scott Hansen
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - Michael A Bender
- Department of Pediatrics, University of Washington, 1959 NE Pacific St, Health Sciences Building, Seattle, WA Box 356320, 98195 USA ; Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109 USA
| | - John Stamatoyannopoulos
- Department of Genome Sciences, University of Washington, Foege Building S-250, 3720 15th Ave NE, Box 355065, Seattle, WA 98195 USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, 1959 NE Pacific Street, Box 357420, Seattle, WA 98195 USA
| |
Collapse
|
126
|
Hoerder-Suabedissen A, Molnár Z. Development, evolution and pathology of neocortical subplate neurons. Nat Rev Neurosci 2015; 16:133-46. [DOI: 10.1038/nrn3915] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
127
|
Bozek K, Wei Y, Yan Z, Liu X, Xiong J, Sugimoto M, Tomita M, Pääbo S, Sherwood CC, Hof PR, Ely JJ, Li Y, Steinhauser D, Willmitzer L, Giavalisco P, Khaitovich P. Organization and evolution of brain lipidome revealed by large-scale analysis of human, chimpanzee, macaque, and mouse tissues. Neuron 2015; 85:695-702. [PMID: 25661180 DOI: 10.1016/j.neuron.2015.01.003] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/28/2014] [Accepted: 12/31/2014] [Indexed: 01/08/2023]
Abstract
Lipids are prominent components of the nervous system. Here we performed a large-scale mass spectrometry-based analysis of the lipid composition of three brain regions as well as kidney and skeletal muscle of humans, chimpanzees, rhesus macaques, and mice. The human brain shows the most distinct lipid composition: 76% of 5,713 lipid compounds examined in our study are either enriched or depleted in the human brain. Concentration levels of lipids enriched in the brain evolve approximately four times faster among primates compared with lipids characteristic of non-neural tissues and show further acceleration of change in human neocortical regions but not in the cerebellum. Human-specific concentration changes are supported by human-specific expression changes for corresponding enzymes. These results provide the first insights into the role of lipids in human brain evolution.
Collapse
Affiliation(s)
- Katarzyna Bozek
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, Shanghai 200031, China; Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Yuning Wei
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, Shanghai 200031, China
| | - Zheng Yan
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, Shanghai 200031, China
| | - Xiling Liu
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, Shanghai 200031, China
| | - Jieyi Xiong
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, Shanghai 200031, China
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, 997-0035 Tsuruoka, Yamagata, Japan
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, 997-0035 Tsuruoka, Yamagata, Japan
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John J Ely
- Alamogordo Primate Facility, Holloman AFB, Alamogordo, NM 88330, USA
| | - Yan Li
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Dirk Steinhauser
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Lothar Willmitzer
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Patrick Giavalisco
- Max Planck Institute for Molecular Plant Physiology, Am Mühlenberg 1, 14476 Potsdam, Germany.
| | - Philipp Khaitovich
- CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, 320 Yue Yang Road, Shanghai 200031, China; Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, 04103 Leipzig, Germany; Skoltech Center for Computational and Systems Biology, Skolkovo Institute for Science and Technology, Skolkovo 143025, Russia.
| |
Collapse
|
128
|
Myers EM, Bartlett CW, Machiraju R, Bohland JW. An integrative analysis of regional gene expression profiles in the human brain. Methods 2015; 73:54-70. [DOI: 10.1016/j.ymeth.2014.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/27/2014] [Accepted: 12/06/2014] [Indexed: 10/24/2022] Open
|
129
|
Pfenning AR, Hara E, Whitney O, Rivas MV, Wang R, Roulhac PL, Howard JT, Wirthlin M, Lovell PV, Ganapathy G, Mouncastle J, Moseley MA, Thompson JW, Soderblom EJ, Iriki A, Kato M, Gilbert MTP, Zhang G, Bakken T, Bongaarts A, Bernard A, Lein E, Mello CV, Hartemink AJ, Jarvis ED. Convergent transcriptional specializations in the brains of humans and song-learning birds. Science 2014; 346:1256846. [PMID: 25504733 DOI: 10.1126/science.1256846] [Citation(s) in RCA: 299] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
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.
Collapse
Affiliation(s)
- Andreas R Pfenning
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA.
| | - Erina Hara
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - Osceola Whitney
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - Miriam V Rivas
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - Rui Wang
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - Petra L Roulhac
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - Jason T Howard
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - Morgan Wirthlin
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Peter V Lovell
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ganeshkumar Ganapathy
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - Jacquelyn Mouncastle
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA
| | - M Arthur Moseley
- Duke Proteomics and Metabolomics Core Facility, Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - J Will Thompson
- Duke Proteomics and Metabolomics Core Facility, Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Erik J Soderblom
- Duke Proteomics and Metabolomics Core Facility, Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Atsushi Iriki
- Laboratory for Symbolic Cognitive Development, Brain Science Institute, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Masaki Kato
- Laboratory for Symbolic Cognitive Development, Brain Science Institute, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - M Thomas P Gilbert
- Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, 1350 Copenhagen, Denmark. Trace and Environmental DNA Laboratory, Department of Environment and Agriculture, Curtin University, Perth, Western Australia 6102, Australia
| | - Guojie Zhang
- China National GeneBank, BGI-Shenzhen, Shenzhen 518083, China. Centre for Social Evolution, Department of Biology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Trygve Bakken
- Allen Institute for Brain Science, Seattle, WA 98103, USA
| | | | - Amy Bernard
- Allen Institute for Brain Science, Seattle, WA 98103, USA
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, WA 98103, USA
| | - Claudio V Mello
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Erich D Jarvis
- Department of Neurobiology, Howard Hughes Medical Institute, and Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
130
|
Yamawaki N, Borges K, Suter BA, Harris KD, Shepherd GMG. A genuine layer 4 in motor cortex with prototypical synaptic circuit connectivity. eLife 2014; 3:e05422. [PMID: 25525751 PMCID: PMC4290446 DOI: 10.7554/elife.05422] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/18/2014] [Indexed: 12/28/2022] Open
Abstract
The motor cortex (M1) is classically considered an agranular area, lacking a distinct layer 4 (L4). Here, we tested the idea that M1, despite lacking a cytoarchitecturally visible L4, nevertheless possesses its equivalent in the form of excitatory neurons with input–output circuits like those of the L4 neurons in sensory areas. Consistent with this idea, we found that neurons located in a thin laminar zone at the L3/5A border in the forelimb area of mouse M1 have multiple L4-like synaptic connections: excitatory input from thalamus, largely unidirectional excitatory outputs to L2/3 pyramidal neurons, and relatively weak long-range corticocortical inputs and outputs. M1-L4 neurons were electrophysiologically diverse but morphologically uniform, with pyramidal-type dendritic arbors and locally ramifying axons, including branches extending into L2/3. Our findings therefore identify pyramidal neurons in M1 with the expected prototypical circuit properties of excitatory L4 neurons, and question the traditional assumption that motor cortex lacks this layer. DOI:http://dx.doi.org/10.7554/eLife.05422.001 In 1909, a German scientist called Korbinian Brodmann published the first map of the outer layer of the human brain. After staining neurons with a dye and studying the structures of the cells and how they were organized, he realized that he could divide the cortex into 43 numbered regions. Most Brodmann areas can be divided into a number of horizontal layers, with layer 1 being closest to the surface of the brain. Neurons in the different layers form distinct sets of connections, and the relative thickness of the layers has implications for the function carried out by that area. It is thought, for example, that the motor cortex does not have a layer 4, which suggests that the neural circuitry that controls movement differs from that in charge of vision, hearing, and other functions. Yamawaki et al. now challenge this view by providing multiple lines of evidence for the existence of layer 4 in the motor cortex in mice. Neurons at the border between layer 3 and layer 5A in the motor cortex possess many of the same properties as the neurons in layer 4 in sensory cortex. In particular, they receive inputs from a brain region called the thalamus, and send outputs to neurons in layers 2 and 3. Yamawaki et al. go on to characterize some of the properties of the neurons in the putative layer 4 of the motor cortex, finding that they do not look like the specialized ‘stellate’ cells that are found in some other areas of the cortex. Instead, they resemble the ‘pyramidal’ type of neuron that is found in all layers and areas of the cortex. The discovery that the motor cortex is more similar in its circuit connections to other area of the cortex than previously thought has important implications for our understanding of this region of the brain. DOI:http://dx.doi.org/10.7554/eLife.05422.002
Collapse
Affiliation(s)
- Naoki Yamawaki
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Katharine Borges
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Benjamin A Suter
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Kenneth D Harris
- Institute of Neurology, University College London, London, United Kingdom
| | - Gordon M G Shepherd
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, United States
| |
Collapse
|
131
|
Lodato S, Shetty AS, Arlotta P. Cerebral cortex assembly: generating and reprogramming projection neuron diversity. Trends Neurosci 2014; 38:117-25. [PMID: 25529141 DOI: 10.1016/j.tins.2014.11.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/11/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
Abstract
The mammalian cerebral cortex is responsible for the highest levels of associative, cognitive and motor functions. In the central nervous system (CNS) the cortex stands as a prime example of extreme neuronal diversity, broadly classified into excitatory projection neurons (PNs) and inhibitory interneurons (INs). We review here recent progress made in understanding the strategies and mechanisms that shape PN diversity during embryogenesis, and discuss how PN classes may be maintained, postnatally, for the life of the organism. In addition, we consider the intriguing possibility that PNs may be amenable to directed reprogramming of their class-specific features to allow enhanced cortical plasticity in the adult.
Collapse
Affiliation(s)
- Simona Lodato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Ashwin S Shetty
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
132
|
Ha T, Swanson D, Larouche M, Glenn R, Weeden D, Zhang P, Hamre K, Langston M, Phillips C, Song M, Ouyang Z, Chesler E, Duvvurru S, Yordanova R, Cui Y, Campbell K, Ricker G, Phillips C, Homayouni R, Goldowitz D. CbGRiTS: cerebellar gene regulation in time and space. Dev Biol 2014; 397:18-30. [PMID: 25446528 DOI: 10.1016/j.ydbio.2014.09.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 08/23/2014] [Accepted: 09/27/2014] [Indexed: 01/09/2023]
Abstract
The mammalian CNS is one of the most complex biological systems to understand at the molecular level. The temporal information from time series transcriptome analysis can serve as a potent source of associative information between developmental processes and regulatory genes. Here, we introduce a new transcriptome database called, Cerebellar Gene Regulation in Time and Space (CbGRiTS). This dataset is populated with transcriptome data across embryonic and postnatal development from two standard mouse strains, C57BL/6J and DBA/2J, several recombinant inbred lines and cerebellar mutant strains. Users can evaluate expression profiles across cerebellar development in a deep time series with graphical interfaces for data exploration and link-out to anatomical expression databases. We present three analytical approaches that take advantage of specific aspects of the time series for transcriptome analysis. We demonstrate the use of CbGRiTS dataset as a community resource to explore patterns of gene expression and develop hypotheses concerning gene regulatory networks in brain development.
Collapse
Affiliation(s)
- Thomas Ha
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| | - Douglas Swanson
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| | - Matt Larouche
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| | - Randy Glenn
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| | - Dave Weeden
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| | - Peter Zhang
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| | - Kristin Hamre
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Michael Langston
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN, USA
| | - Charles Phillips
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, TN, USA
| | - Mingzhou Song
- Department of Computer Science, New Mexico State University, Las Cruces, NM, USA
| | - Zhengyu Ouyang
- Department of Computer Science, New Mexico State University, Las Cruces, NM, USA
| | | | | | | | - Yan Cui
- Department of Molecular Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kate Campbell
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4
| | - Greg Ricker
- Department of Biology, Bowdoin College, Brunswick, ME, USA
| | - Carey Phillips
- Department of Biology, Bowdoin College, Brunswick, ME, USA
| | - Ramin Homayouni
- Bioinformatics Program, Department of Biology, University of Memphis, Memphis, TN, USA
| | - Dan Goldowitz
- Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia, 950 West 28th Avenue, Vancouver, BC, Canada V5Z 4H4.
| |
Collapse
|
133
|
Horwitz GD. What studies of macaque monkeys have told us about human color vision. Neuroscience 2014; 296:110-5. [PMID: 25445192 DOI: 10.1016/j.neuroscience.2014.10.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/29/2014] [Accepted: 10/01/2014] [Indexed: 11/29/2022]
Abstract
Animal models are a necessary component of systems neuroscience research. Determining which animal model to use for a given study involves a complicated calculus. Some experimental manipulations are easily made in some animal models but impossible in others. Some animal models are similar to humans with respect to particular scientific questions, and others are less so. In this review, I discuss work done in my laboratory to investigate the neural mechanisms of color vision in the rhesus macaque. The emphasis is on the strengths of the macaque model, but shortcomings are also discussed.
Collapse
Affiliation(s)
- G D Horwitz
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States; Washington National Primate Research Center, Seattle, WA, United States.
| |
Collapse
|
134
|
Bohland JW, Myers EM, Kim E. An informatics approach to integrating genetic and neurological data in speech and language neuroscience. Neuroinformatics 2014; 12:39-62. [PMID: 23949335 DOI: 10.1007/s12021-013-9201-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
A number of heritable disorders impair the normal development of speech and language processes and occur in large numbers within the general population. While candidate genes and loci have been identified, the gap between genotype and phenotype is vast, limiting current understanding of the biology of normal and disordered processes. This gap exists not only in our scientific knowledge, but also in our research communities, where genetics researchers and speech, language, and cognitive scientists tend to operate independently. Here we describe a web-based, domain-specific, curated database that represents information about genotype-phenotype relations specific to speech and language disorders, as well as neuroimaging results demonstrating focal brain differences in relevant patients versus controls. Bringing these two distinct data types into a common database ( http://neurospeech.org/sldb ) is a first step toward bringing molecular level information into cognitive and computational theories of speech and language function. One bridge between these data types is provided by densely sampled profiles of gene expression in the brain, such as those provided by the Allen Brain Atlases. Here we present results from exploratory analyses of human brain gene expression profiles for genes implicated in speech and language disorders, which are annotated in our database. We then discuss how such datasets can be useful in the development of computational models that bridge levels of analysis, necessary to provide a mechanistic understanding of heritable language disorders. We further describe our general approach to information integration, discuss important caveats and considerations, and offer a specific but speculative example based on genes implicated in stuttering and basal ganglia function in speech motor control.
Collapse
Affiliation(s)
- Jason W Bohland
- Departments of Health Sciences and Speech, Language, and Hearing Sciences, Boston University, 635 Commonwealth Ave, Room 403, Boston, MA, 02215, USA,
| | | | | |
Collapse
|
135
|
Balaram P, Kaas JH. Towards a unified scheme of cortical lamination for primary visual cortex across primates: insights from NeuN and VGLUT2 immunoreactivity. Front Neuroanat 2014; 8:81. [PMID: 25177277 PMCID: PMC4133926 DOI: 10.3389/fnana.2014.00081] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 07/23/2014] [Indexed: 12/02/2022] Open
Abstract
Primary visual cortex (V1) is clearly distinguishable from other cortical areas by its distinctive pattern of neocortical lamination across mammalian species. In some mammals, primates in particular, the layers of V1 are further divided into a number of sublayers based on their anatomical and functional characteristics. While these sublayers are easily recognizable across a range of primates, the exact number of divisions in each layer and their relative position within the depth of V1 has been inconsistently reported, largely due to conflicting schemes of nomenclature for the V1 layers. This conflict centers on the definition of layer 4 in primate V1, and the subdivisions of layer 4 that can be consistently identified across primate species. Brodmann’s (1909) laminar scheme for V1 delineates three subdivisions of layer 4 in primates, based on cellular morphology and geniculate inputs in anthropoid monkeys. In contrast, Hässler’s (1967) laminar scheme delineates a single layer 4 and multiple subdivisions of layer 3, based on comparisons of V1 lamination across the primate lineage. In order to clarify laminar divisions in primate visual cortex, we performed NeuN and VGLUT2 immunohistochemistry in V1 of chimpanzees, Old World macaque monkeys, New World squirrel, owl, and marmoset monkeys, prosimian galagos and mouse lemurs, and non-primate, but highly visual, tree shrews. By comparing the laminar divisions identified by each method across species, we find that Hässler’s (1967) laminar scheme for V1 provides a more consistent representation of neocortical layers across all primates, including humans, and facilitates comparisons of V1 lamination with non-primate species. These findings, along with many others, support the consistent use of Hässler’s laminar scheme in V1 research.
Collapse
Affiliation(s)
- Pooja Balaram
- Laboratory of Jon Kaas, Department of Psychology, Vanderbilt University Nashville, TN, USA
| | - Jon H Kaas
- Laboratory of Jon Kaas, Department of Psychology, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
136
|
Markov NT, Vezoli J, Chameau P, Falchier A, Quilodran R, Huissoud C, Lamy C, Misery P, Giroud P, Ullman S, Barone P, Dehay C, Knoblauch K, Kennedy H. Anatomy of hierarchy: feedforward and feedback pathways in macaque visual cortex. J Comp Neurol 2014; 522:225-59. [PMID: 23983048 PMCID: PMC4255240 DOI: 10.1002/cne.23458] [Citation(s) in RCA: 453] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 04/10/2013] [Accepted: 08/14/2013] [Indexed: 12/18/2022]
Abstract
The laminar location of the cell bodies and terminals of interareal connections determines the hierarchical structural organization of the cortex and has been intensively studied. However, we still have only a rudimentary understanding of the connectional principles of feedforward (FF) and feedback (FB) pathways. Quantitative analysis of retrograde tracers was used to extend the notion that the laminar distribution of neurons interconnecting visual areas provides an index of hierarchical distance (percentage of supragranular labeled neurons [SLN]). We show that: 1) SLN values constrain models of cortical hierarchy, revealing previously unsuspected areal relations; 2) SLN reflects the operation of a combinatorial distance rule acting differentially on sets of connections between areas; 3) Supragranular layers contain highly segregated bottom-up and top-down streams, both of which exhibit point-to-point connectivity. This contrasts with the infragranular layers, which contain diffuse bottom-up and top-down streams; 4) Cell filling of the parent neurons of FF and FB pathways provides further evidence of compartmentalization; 5) FF pathways have higher weights, cross fewer hierarchical levels, and are less numerous than FB pathways. Taken together, the present results suggest that cortical hierarchies are built from supra- and infragranular counterstreams. This compartmentalized dual counterstream organization allows point-to-point connectivity in both bottom-up and top-down directions.
Collapse
Affiliation(s)
- Nikola T Markov
- Stem Cell and Brain Research Institute, INSERM U846, 69500, Bron, France; Université de Lyon, Université Lyon I, 69003, Lyon, France; Department of Neurobiology, Yale University School of Medicine, New Haven, Connecticut, 06520-8001, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Kumamoto T, Hanashima C. Neuronal subtype specification in establishing mammalian neocortical circuits. Neurosci Res 2014; 86:37-49. [PMID: 25019611 DOI: 10.1016/j.neures.2014.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/21/2014] [Accepted: 06/23/2014] [Indexed: 11/28/2022]
Abstract
The functional integrity of the neocortical circuit relies on the precise production of diverse neuron populations and their assembly during development. In recent years, extensive progress has been made in the understanding of the mechanisms that control differentiation of each neuronal type within the neocortex. In this review, we address how the elaborate neocortical cytoarchitecture is established from a simple neuroepithelium based on recent studies examining the spatiotemporal mechanisms of neuronal subtype specification. We further discuss the critical events that underlie the conversion of the stem amniotes cerebrum to a mammalian-type neocortex, and extend these key findings in the light of mammalian evolution to understand how the neocortex in humans evolved from common ancestral mammals.
Collapse
Affiliation(s)
- Takuma Kumamoto
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Carina Hanashima
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Department of Biology, Graduate School of Science, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
138
|
Homman-Ludiye J, Bourne JA. Mapping arealisation of the visual cortex of non-primate species: lessons for development and evolution. Front Neural Circuits 2014; 8:79. [PMID: 25071460 PMCID: PMC4081835 DOI: 10.3389/fncir.2014.00079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023] Open
Abstract
The integration of the visual stimulus takes place at the level of the neocortex, organized in anatomically distinct and functionally unique areas. Primates, including humans, are heavily dependent on vision, with approximately 50% of their neocortical surface dedicated to visual processing and possess many more visual areas than any other mammal, making them the model of choice to study visual cortical arealisation. However, in order to identify the mechanisms responsible for patterning the developing neocortex, specifying area identity as well as elucidate events that have enabled the evolution of the complex primate visual cortex, it is essential to gain access to the cortical maps of alternative species. To this end, species including the mouse have driven the identification of cellular markers, which possess an area-specific expression profile, the development of new tools to label connections and technological advance in imaging techniques enabling monitoring of cortical activity in a behaving animal. In this review we present non-primate species that have contributed to elucidating the evolution and development of the visual cortex. We describe the current understanding of the mechanisms supporting the establishment of areal borders during development, mainly gained in the mouse thanks to the availability of genetically modified lines but also the limitations of the mouse model and the need for alternate species.
Collapse
Affiliation(s)
- Jihane Homman-Ludiye
- Bourne Group, Australian Regenerative Medicine Institute, Monash University Clayton, VIC, Australia
| | - James A Bourne
- Bourne Group, Australian Regenerative Medicine Institute, Monash University Clayton, VIC, Australia
| |
Collapse
|
139
|
Kawasaki H. Molecular investigations of the brain of higher mammals using gyrencephalic carnivore ferrets. Neurosci Res 2014; 86:59-65. [PMID: 24983876 DOI: 10.1016/j.neures.2014.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 11/17/2022]
Abstract
The brains of mammals such as carnivores and primates contain developed structures not found in the brains of mice. Uncovering the physiological importance, developmental mechanisms and evolution of these structures using carnivores and primates would greatly contribute to our understanding of the human brain and its diseases. Although the anatomical and physiological properties of the brains of carnivores and primates have been intensively examined, molecular investigations are still limited. Recently, genetic techniques that can be applied to carnivores and primates have been explored, and molecules whose expression patterns correspond to these structures were reported. Furthermore, to investigate the functional importance of these molecules, rapid and efficient genetic manipulation methods were established by applying electroporation to gyrencephalic carnivore ferrets. In this article, I review recent advances in molecular investigations of the brains of carnivores and primates, mainly focusing on ferrets (Mustela putorius furo).
Collapse
Affiliation(s)
- Hiroshi Kawasaki
- Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan; Brain/Liver Interface Medicine Research Center, Kanazawa University, Ishikawa 920-8640, Japan.
| |
Collapse
|
140
|
Hrvoj-Mihic B, Marchetto MCN, Gage FH, Semendeferi K, Muotri AR. Novel tools, classic techniques: evolutionary studies using primate pluripotent stem cells. Biol Psychiatry 2014; 75:929-35. [PMID: 24041506 DOI: 10.1016/j.biopsych.2013.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 07/25/2013] [Accepted: 08/06/2013] [Indexed: 11/25/2022]
Abstract
Recent applications of genomic tools on the analysis of alterations unique to our species coupled with a growing number of neuroanatomical studies across primates provide an unprecedented opportunity to compile different levels of human brain evolution into a complex whole. Applications of induced pluripotent stem cell (iPSC) technology, capable of reprogramming somatic tissue of different species and generating species-specific neuronal phenotypes, for the first time offer an opportunity to test specific evolutionary hypotheses in a field of inquiry that has been long plagued by the limited availability of research specimens. In this review, we will focus specifically on the experimental role of iPSC technology as applied to the analysis of neocortical pyramidal neurons. Pyramidal neurons emerge as particularly suitable for testing evolutionary scenarios, since they form the most common morphological class of neurons in the cortex, display morphological variations across different cortical areas and cortical layers that appear species-specific, and express unique molecular signatures. Human and nonhuman primate iPSC-derived neurons may represent a unique biological resource to elucidate the phenotypic differences between humans and other hominids. As the typical morphology of pyramidal neurons tends to be compromised in neurological disorders, application of iPSC technology to the analysis of pyramidal neurons could not only bring new insights into human adaptation but also offer opportunities to link biomedical research with studies of the origins of the human species.
Collapse
Affiliation(s)
- Branka Hrvoj-Mihic
- Department of Anthropology; School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, University of California San Diego
| | - Maria C N Marchetto
- Laboratory of Genetics (MCNM, FHG), The Salk Institute for Biological Studies
| | - Fred H Gage
- Laboratory of Genetics (MCNM, FHG), The Salk Institute for Biological Studies; Center for Academic Research and Training in Anthropogeny
| | - Katerina Semendeferi
- Department of Anthropology; Center for Academic Research and Training in Anthropogeny; Neuroscience Graduate Program, University of California San Diego, La Jolla, California
| | - Alysson R Muotri
- School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, Department of Cellular & Molecular Medicine, Stem Cell Program, University of California San Diego; Center for Academic Research and Training in Anthropogeny; Neuroscience Graduate Program, University of California San Diego, La Jolla, California.
| |
Collapse
|
141
|
Molnár Z, Kaas JH, de Carlos JA, Hevner RF, Lein E, Němec P. Evolution and development of the mammalian cerebral cortex. BRAIN, BEHAVIOR AND EVOLUTION 2014; 83:126-39. [PMID: 24776993 PMCID: PMC4440552 DOI: 10.1159/000357753] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/03/2013] [Indexed: 12/20/2022]
Abstract
Comparative developmental studies of the mammalian brain can identify key changes that can generate the diverse structures and functions of the brain. We have studied how the neocortex of early mammals became organized into functionally distinct areas, and how the current level of cortical cellular and laminar specialization arose from the simpler premammalian cortex. We demonstrate the neocortical organization in early mammals, which helps to elucidate how the large, complex human brain evolved from a long line of ancestors. The radial and tangential enlargement of the cortex was driven by changes in the patterns of cortical neurogenesis, including alterations in the proportions of distinct progenitor types. Some cortical cell populations travel to the cortex through tangential migration whereas others migrate radially. A number of recent studies have begun to characterize the chick, mouse and human and nonhuman primate cortical transcriptome to help us understand how gene expression relates to the development and anatomical and functional organization of the adult neocortex. Although all mammalian forms share the basic layout of cortical areas, the areal proportions and distributions are driven by distinct evolutionary pressures acting on sensory and motor experiences during the individual ontogenies.
Collapse
Affiliation(s)
- Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | | | | | | | |
Collapse
|
142
|
Miller JA, Ding SL, Sunkin SM, Smith KA, Ng L, Szafer A, Ebbert A, Riley ZL, Royall JJ, Aiona K, Arnold JM, Bennet C, Bertagnolli D, Brouner K, Butler S, Caldejon S, Carey A, Cuhaciyan C, Dalley RA, Dee N, Dolbeare TA, Facer BAC, Feng D, Fliss TP, Gee G, Goldy J, Gourley L, Gregor BW, Gu G, Howard RE, Jochim JM, Kuan CL, Lau C, Lee CK, Lee F, Lemon TA, Lesnar P, McMurray B, Mastan N, Mosqueda N, Naluai-Cecchini T, Ngo NK, Nyhus J, Oldre A, Olson E, Parente J, Parker PD, Parry SE, Stevens A, Pletikos M, Reding M, Roll K, Sandman D, Sarreal M, Shapouri S, Shapovalova NV, Shen EH, Sjoquist N, Slaughterbeck CR, Smith M, Sodt AJ, Williams D, Zöllei L, Fischl B, Gerstein MB, Geschwind DH, Glass IA, Hawrylycz MJ, Hevner RF, Huang H, Jones AR, Knowles JA, Levitt P, Phillips JW, Sestan N, Wohnoutka P, Dang C, Bernard A, Hohmann JG, Lein ES. Transcriptional landscape of the prenatal human brain. Nature 2014; 508:199-206. [PMID: 24695229 PMCID: PMC4105188 DOI: 10.1038/nature13185] [Citation(s) in RCA: 917] [Impact Index Per Article: 83.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 02/26/2014] [Indexed: 12/21/2022]
Abstract
The anatomical and functional architecture of the human brain is largely determined by prenatal transcriptional processes. We describe an anatomically comprehensive atlas of 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 postmitotic 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 human-expanded outer subventricular zones. Both germinal and postmitotic cortical layers exhibit fronto-temporal gradients, with particular enrichment in 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.
Collapse
Affiliation(s)
- Jeremy A Miller
- 1] Allen Institute for Brain Science, Seattle, Washington 98103, USA [2]
| | - Song-Lin Ding
- 1] Allen Institute for Brain Science, Seattle, Washington 98103, USA [2]
| | - Susan M Sunkin
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Kimberly A Smith
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Lydia Ng
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Aaron Szafer
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Amanda Ebbert
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Zackery L Riley
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Joshua J Royall
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Kaylynn Aiona
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - James M Arnold
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Crissa Bennet
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | | | - Krissy Brouner
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Stephanie Butler
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Shiella Caldejon
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Anita Carey
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | | | - Rachel A Dalley
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Nick Dee
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Tim A Dolbeare
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | | | - David Feng
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Tim P Fliss
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Garrett Gee
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Lindsey Gourley
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | | | - Guangyu Gu
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Robert E Howard
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Jayson M Jochim
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Chihchau L Kuan
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Christopher Lau
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Chang-Kyu Lee
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Felix Lee
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Tracy A Lemon
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Phil Lesnar
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Bergen McMurray
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Naveed Mastan
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Nerick Mosqueda
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Theresa Naluai-Cecchini
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, 1959 North East Pacific Street, Box 356320, Seattle, Washington 98195, USA
| | - Nhan-Kiet Ngo
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Julie Nyhus
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Aaron Oldre
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Eric Olson
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Jody Parente
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Patrick D Parker
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Sheana E Parry
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Allison Stevens
- 1] Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA [2] Computer Science and AI Lab, MIT, Cambridge, Massachusetts 02139, USA
| | - Mihovil Pletikos
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Melissa Reding
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Kate Roll
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - David Sandman
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Melaine Sarreal
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Sheila Shapouri
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | | | - Elaine H Shen
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Nathan Sjoquist
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | | | - Michael Smith
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Andy J Sodt
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Derric Williams
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Lilla Zöllei
- Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | - Bruce Fischl
- 1] Department of Radiology, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA [2] Computer Science and AI Lab, MIT, Cambridge, Massachusetts 02139, USA
| | - Mark B Gerstein
- 1] Program in Computational Biology and Bioinformatics, Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut 06520, USA [2] Department of Computer Science, Yale University, New Haven, Connecticut 06520, USA
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology and Semel Institute David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
| | - Ian A Glass
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, 1959 North East Pacific Street, Box 356320, Seattle, Washington 98195, USA
| | | | - Robert F Hevner
- 1] Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101, USA [2] Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98105, USA
| | - Hao Huang
- Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Allan R Jones
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - James A Knowles
- Zilkha Neurogenetic Institute, and Department of Psychiatry, University of Southern California, Los Angeles, California 90033, USA
| | - Pat Levitt
- 1] Department of Pediatrics, Children's Hospital, Los Angeles, California 90027, USA [2] Keck School of Medicine, University of Southern California, Los Angeles, California 90089, USA
| | - John W Phillips
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Nenad Sestan
- Department of Neurobiology and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, USA
| | - Paul Wohnoutka
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Chinh Dang
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Amy Bernard
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - John G Hohmann
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| | - Ed S Lein
- Allen Institute for Brain Science, Seattle, Washington 98103, USA
| |
Collapse
|
143
|
Role of radial glial cells in cerebral cortex folding. Curr Opin Neurobiol 2014; 27:39-46. [PMID: 24632307 DOI: 10.1016/j.conb.2014.02.007] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/22/2014] [Accepted: 02/06/2014] [Indexed: 11/24/2022]
Abstract
Radial glial cells play key roles during cerebral cortex development, as primary stem and progenitor cells giving rise-directly or indirectly-to neurons and glia, but also acting as scaffold for the cerebral cortex architecture and migrating neurons. Recent work led to the discovery of novel types of radial glial cells with key roles in gyrification, the folding of the mammalian cerebral cortex in phylogeny and ontogeny. Here we summarize the cellular and molecular basis of this fascinating process allowing the expansion of the mammalian cerebral cortex with all its functional consequences.
Collapse
|
144
|
DNA methylation and methyl-binding proteins control differential gene expression in distinct cortical areas of macaque monkey. J Neurosci 2014; 33:19704-14. [PMID: 24336734 DOI: 10.1523/jneurosci.2355-13.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Distinct anatomical regions of the neocortex subserve different sensory modalities and neuronal integration functions, but mechanisms for these regional specializations remain elusive. Involvement of epigenetic mechanisms for such specialization through the spatiotemporal regulation of gene expression is an intriguing possibility. Here we examined whether epigenetic mechanisms might play a role in the selective gene expression in the association areas (AAs) and the primary visual cortex (V1) in macaque neocortex. By analyzing the two types of area-selective gene promoters that we previously identified, we found a striking difference of DNA methylation between these promoters, i.e., hypermethylation in AA-selective gene promoters and hypomethylation in V1-selective ones. Methylation levels of promoters of each area-selective gene showed no areal difference, but a specific methyl-binding protein (MBD4) was enriched in the AAs, in correspondence with expression patterns of AA-selective genes. MBD4 expression was mainly observed in neurons. MBD4 specifically bound to and activated the AA-selective genes both in vitro and in vivo. Our results demonstrate that methylation in the promoters and specific methyl-binding proteins play an important role in the area-selective gene expression profiles in the primate neocortex.
Collapse
|
145
|
Goel P, Kuceyeski A, LoCastro E, Raj A. Spatial patterns of genome-wide expression profiles reflect anatomic and fiber connectivity architecture of healthy human brain. Hum Brain Mapp 2014; 35:4204-18. [PMID: 24677576 DOI: 10.1002/hbm.22471] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 11/30/2013] [Accepted: 01/06/2014] [Indexed: 11/07/2022] Open
Abstract
Unraveling the relationship between molecular signatures in the brain and their functional, architectonic, and anatomic correlates is an important neuroscientific goal. It is still not well understood whether the diversity demonstrated by histological studies in the human brain is reflected in the spatial patterning of whole brain transcriptional profiles. Using genome-wide maps of transcriptional distribution of the human brain by the Allen Brain Institute, we test the hypothesis that gene expression profiles are specific to anatomically described brain regions. In this work, we demonstrate that this is indeed the case by showing that gene similarity clusters appear to respect conventional basal-cortical and caudal-rostral gradients. To fully investigate the causes of this observed spatial clustering, we test a connectionist hypothesis that states that the spatial patterning of gene expression in the brain is simply reflective of the fiber tract connectivity between brain regions. We find that although gene expression and structural connectivity are not determined by each other, they do influence each other with a high statistical significance. This implies that spatial diversity of gene expressions is a result of mainly location-specific features but is influenced by neuronal connectivity, such that like cellular species preferentially connects with like cells.
Collapse
Affiliation(s)
- Pragya Goel
- Department of Computer Science, Cornell University, Ithaca, New York
| | | | | | | |
Collapse
|
146
|
Parikshak NN, Luo R, Zhang A, Won H, Lowe JK, Chandran V, Horvath S, Geschwind DH. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 2014; 155:1008-21. [PMID: 24267887 DOI: 10.1016/j.cell.2013.10.031] [Citation(s) in RCA: 749] [Impact Index Per Article: 68.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 07/31/2013] [Accepted: 10/03/2013] [Indexed: 01/09/2023]
Abstract
Genetic studies have identified dozens of autism spectrum disorder (ASD) susceptibility genes, raising two critical questions: (1) do these genetic loci converge on specific biological processes, and (2) where does the phenotypic specificity of ASD arise, given its genetic overlap with intellectual disability (ID)? To address this, we mapped ASD and ID risk genes onto coexpression networks representing developmental trajectories and transcriptional profiles representing fetal and adult cortical laminae. ASD genes tightly coalesce in modules that implicate distinct biological functions during human cortical development, including early transcriptional regulation and synaptic development. Bioinformatic analyses suggest that translational regulation by FMRP and transcriptional coregulation by common transcription factors connect these processes. At a circuit level, ASD genes are enriched in superficial cortical layers and glutamatergic projection neurons. Furthermore, we show that the patterns of ASD and ID risk genes are distinct, providing a biological framework for further investigating the pathophysiology of ASD.
Collapse
Affiliation(s)
- Neelroop N Parikshak
- Program in Neurobehavioral Genetics, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Interdepartmental Program in Neuroscience, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
147
|
Boudreau RL, Jiang P, Gilmore BL, Spengler RM, Tirabassi R, Nelson JA, Ross CA, Xing Y, Davidson BL. Transcriptome-wide discovery of microRNA binding sites in human brain. Neuron 2014; 81:294-305. [PMID: 24389009 DOI: 10.1016/j.neuron.2013.10.062] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2013] [Indexed: 12/15/2022]
Abstract
The orchestration of brain function requires complex gene regulatory networks that are modulated, in part, by microRNAs (miRNAs). These noncoding RNAs associate with argonaute (Ago) proteins in order to direct posttranscriptional gene suppression via base pairing with target transcripts. In order to better understand how miRNAs contribute to human-specialized brain processes and neurological phenotypes, identifying their targets is of paramount importance. Here, we address the latter by profiling Ago2:RNA interactions using HITS-CLIP to generate a transcriptome-wide map of miRNA binding sites in human brain. We uncovered ∼ 7,000 stringent Ago2 binding sites that are highly enriched for conserved sequences corresponding to abundant brain miRNAs. This interactome points to functional miRNA:target pairs across >3,000 genes and represents a valuable resource for accelerating our understanding of miRNA functions in brain. We demonstrate the utility of this map for exploring clinically relevant miRNA binding sites that may facilitate the translation of genetic studies of complex neuropsychiatric diseases into therapeutics.
Collapse
Affiliation(s)
- Ryan L Boudreau
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Peng Jiang
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Brian L Gilmore
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ryan M Spengler
- Program in Molecular and Cellular Biology, University of Iowa, Iowa City, IA 52242, USA
| | - Rebecca Tirabassi
- Vaccine and Gene Therapy Institute, Oregon Health & Sciences University, Beaverton, OR 97006, USA
| | - Jay A Nelson
- Vaccine and Gene Therapy Institute, Oregon Health & Sciences University, Beaverton, OR 97006, USA
| | - Christopher A Ross
- Division of Neurobiology; Departments of Psychiatry, Neurology Neuroscience, and Pharmacology; and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yi Xing
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Beverly L Davidson
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA; Department of Neurology, University of Iowa, Iowa City, IA 52242, USA; Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA 52242, USA; Program in Molecular and Cellular Biology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
148
|
Agis-Balboa RC, Fischer A. Generating new neurons to circumvent your fears: the role of IGF signaling. Cell Mol Life Sci 2014; 71:21-42. [PMID: 23543251 PMCID: PMC11113432 DOI: 10.1007/s00018-013-1316-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 02/12/2013] [Accepted: 03/04/2013] [Indexed: 12/13/2022]
Abstract
Extinction of fear memory is a particular form of cognitive function that is of special interest because of its involvement in the treatment of anxiety and mood disorders. Based on recent literature and our previous findings (EMBO J 30(19):4071-4083, 2011), we propose a new hypothesis that implies a tight relationship among IGF signaling, adult hippocampal neurogenesis and fear extinction. Our proposed model suggests that fear extinction-induced IGF2/IGFBP7 signaling promotes the survival of neurons at 2-4 weeks old that would participate in the discrimination between the original fear memory trace and the new safety memory generated during fear extinction. This is also called "pattern separation", or the ability to distinguish similar but different cues (e.g., context). To understand the molecular mechanisms underlying fear extinction is therefore of great clinical importance.
Collapse
Affiliation(s)
- R C Agis-Balboa
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Grisebach Str. 5, 37077, Göttingen, Germany,
| | | |
Collapse
|
149
|
Abstract
The human cerebral cortex is generally considered the most complex organ, and is the structure that we hold responsible for the repertoire of behavior that distinguishes us from our closest living and extinct relatives. At a recent Company of Biologists Workshop, ‘Evolution of the Human Neocortex: How Unique Are We?’ held in September 2013, researchers considered new information from the fields of developmental biology, genetics, genomics, molecular biology and ethology to understand unique features of the human cerebral cortex and their developmental and evolutionary origin.
Collapse
Affiliation(s)
- Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Alex Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, 35 Medical Center Way, San Francisco, CA 94143, USA
| |
Collapse
|
150
|
Markov NT, Ercsey-Ravasz M, Van Essen DC, Knoblauch K, Toroczkai Z, Kennedy H. Cortical high-density counterstream architectures. Science 2013; 342:1238406. [PMID: 24179228 DOI: 10.1126/science.1238406] [Citation(s) in RCA: 381] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Small-world networks provide an appealing description of cortical architecture owing to their capacity for integration and segregation combined with an economy of connectivity. Previous reports of low-density interareal graphs and apparent small-world properties are challenged by data that reveal high-density cortical graphs in which economy of connections is achieved by weight heterogeneity and distance-weight correlations. These properties define a model that predicts many binary and weighted features of the cortical network including a core-periphery, a typical feature of self-organizing information processing systems. Feedback and feedforward pathways between areas exhibit a dual counterstream organization, and their integration into local circuits constrains cortical computation. Here, we propose a bow-tie representation of interareal architecture derived from the hierarchical laminar weights of pathways between the high-efficiency dense core and periphery.
Collapse
Affiliation(s)
- Nikola T Markov
- Stem cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lépine, 69500 Bron, France.,Université de Lyon, Université Lyon I, 69003 Lyon, France.,Yale University, Department of Neurobiology, New Haven, CT 06520, USA
| | | | - David C Van Essen
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Kenneth Knoblauch
- Stem cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lépine, 69500 Bron, France.,Université de Lyon, Université Lyon I, 69003 Lyon, France
| | - Zoltán Toroczkai
- Department of Physics and Interdisciplinary Center for Network Science and Applications, University of Notre Dame, Notre Dame, IN 46556, USA.,Max Planck Institute for the Physics of Complex Systems, 01187 Dresden, Germany
| | - Henry Kennedy
- Stem cell and Brain Research Institute, INSERM U846, 18 Avenue Doyen Lépine, 69500 Bron, France.,Université de Lyon, Université Lyon I, 69003 Lyon, France
| |
Collapse
|