1
|
Liu Z, Feng Z, Liu G, Li A, Gong H, Yang X, Li X. A complementary approach for neocortical cytoarchitecture inspection with cellular resolution imaging at whole brain scale. Front Neuroanat 2024; 18:1388084. [PMID: 38846539 PMCID: PMC11153794 DOI: 10.3389/fnana.2024.1388084] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/26/2024] [Indexed: 06/09/2024] Open
Abstract
Cytoarchitecture, the organization of cells within organs and tissues, serves as a crucial anatomical foundation for the delineation of various regions. It enables the segmentation of the cortex into distinct areas with unique structural and functional characteristics. While traditional 2D atlases have focused on cytoarchitectonic mapping of cortical regions through individual sections, the intricate cortical gyri and sulci demands a 3D perspective for unambiguous interpretation. In this study, we employed fluorescent micro-optical sectioning tomography to acquire architectural datasets of the entire macaque brain at a resolution of 0.65 μm × 0.65 μm × 3 μm. With these volumetric data, the cortical laminar textures were remarkably presented in appropriate view planes. Additionally, we established a stereo coordinate system to represent the cytoarchitectonic information as surface-based tomograms. Utilizing these cytoarchitectonic features, we were able to three-dimensionally parcel the macaque cortex into multiple regions exhibiting contrasting architectural patterns. The whole-brain analysis was also conducted on mice that clearly revealed the presence of barrel cortex and reflected biological reasonability of this method. Leveraging these high-resolution continuous datasets, our method offers a robust tool for exploring the organizational logic and pathological mechanisms of the brain's 3D anatomical structure.
Collapse
Affiliation(s)
- Zhixiang Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao Feng
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Guangcai Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
| | - Anan Li
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Hui Gong
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Xiaoquan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, MoE Key Laboratory for Biomedical Photonics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| | - Xiangning Li
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| |
Collapse
|
2
|
Molnár Z, Kwan KY. Development and Evolution of Thalamocortical Connectivity. Cold Spring Harb Perspect Biol 2024; 16:a041503. [PMID: 38167425 PMCID: PMC10759993 DOI: 10.1101/cshperspect.a041503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Conscious perception in mammals depends on precise circuit connectivity between cerebral cortex and thalamus; the evolution and development of these structures are closely linked. During the wiring of reciprocal thalamus-cortex connections, thalamocortical axons (TCAs) first navigate forebrain regions that had undergone substantial evolutionary modifications. In particular, the organization of the pallial-subpallial boundary (PSPB) diverged significantly between mammals, reptiles, and birds. In mammals, transient cell populations in internal capsule and early corticofugal projections from subplate neurons closely interact with TCAs to guide pathfinding through ventral forebrain and PSPB crossing. Prior to thalamocortical axon arrival, cortical areas are initially patterned by intrinsic genetic factors. Thalamocortical axons then innervate cortex in a topographically organized manner to enable sensory input to refine cortical arealization. Here, we review the mechanisms underlying the guidance of thalamocortical axons across forebrain boundaries, the implications of PSPB evolution for thalamocortical axon pathfinding, and the reciprocal influence between thalamus and cortex during development.
Collapse
Affiliation(s)
- Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Kenneth Y Kwan
- Michigan Neuroscience Institute (MNI), Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
3
|
Fujita T, Aoki N, Mori C, Homma KJ, Yamaguchi S. Molecular biology of serotonergic systems in avian brains. Front Mol Neurosci 2023; 16:1226645. [PMID: 37538316 PMCID: PMC10394247 DOI: 10.3389/fnmol.2023.1226645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a phylogenetically conserved neurotransmitter and modulator. Neurons utilizing serotonin have been identified in the central nervous systems of all vertebrates. In the central serotonergic system of vertebrate species examined so far, serotonergic neurons have been confirmed to exist in clusters in the brainstem. Although many serotonin-regulated cognitive, behavioral, and emotional functions have been elucidated in mammals, equivalents remain poorly understood in non-mammalian vertebrates. The purpose of this review is to summarize current knowledge of the anatomical organization and molecular features of the avian central serotonergic system. In addition, selected key functions of serotonin are briefly reviewed. Gene association studies between serotonergic system related genes and behaviors in birds have elucidated that the serotonergic system is involved in the regulation of behavior in birds similar to that observed in mammals. The widespread distribution of serotonergic modulation in the central nervous system and the evolutionary conservation of the serotonergic system provide a strong foundation for understanding and comparing the evolutionary continuity of neural circuits controlling corresponding brain functions within vertebrates. The main focus of this review is the chicken brain, with this type of poultry used as a model bird. The chicken is widely used not only as a model for answering questions in developmental biology and as a model for agriculturally useful breeding, but also in research relating to cognitive, behavioral, and emotional processes. In addition to a wealth of prior research on the projection relationships of avian brain regions, detailed subdivision similarities between avian and mammalian brains have recently been identified. Therefore, identifying the neural circuits modulated by the serotonergic system in avian brains may provide an interesting opportunity for detailed comparative studies of the function of serotonergic systems in mammals.
Collapse
Affiliation(s)
- Toshiyuki Fujita
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Naoya Aoki
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Chihiro Mori
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Koichi J. Homma
- Department of Molecular Biology, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| | - Shinji Yamaguchi
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences, Teikyo University, Tokyo, Japan
| |
Collapse
|
4
|
Salamon I, Rasin MR. Evolution of the Neocortex Through RNA-Binding Proteins and Post-transcriptional Regulation. Front Neurosci 2022; 15:803107. [PMID: 35082597 PMCID: PMC8784817 DOI: 10.3389/fnins.2021.803107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
The human neocortex is undoubtedly considered a supreme accomplishment in mammalian evolution. It features a prenatally established six-layered structure which remains plastic to the myriad of changes throughout an organism’s lifetime. A fundamental feature of neocortical evolution and development is the abundance and diversity of the progenitor cell population and their neuronal and glial progeny. These evolutionary upgrades are partially enabled due to the progenitors’ higher proliferative capacity, compartmentalization of proliferative regions, and specification of neuronal temporal identities. The driving force of these processes may be explained by temporal molecular patterning, by which progenitors have intrinsic capacity to change their competence as neocortical neurogenesis proceeds. Thus, neurogenesis can be conceptualized along two timescales of progenitors’ capacity to (1) self-renew or differentiate into basal progenitors (BPs) or neurons or (2) specify their fate into distinct neuronal and glial subtypes which participate in the formation of six-layers. Neocortical development then proceeds through sequential phases of proliferation, differentiation, neuronal migration, and maturation. Temporal molecular patterning, therefore, relies on the precise regulation of spatiotemporal gene expression. An extensive transcriptional regulatory network is accompanied by post-transcriptional regulation that is frequently mediated by the regulatory interplay between RNA-binding proteins (RBPs). RBPs exhibit important roles in every step of mRNA life cycle in any system, from splicing, polyadenylation, editing, transport, stability, localization, to translation (protein synthesis). Here, we underscore the importance of RBP functions at multiple time-restricted steps of early neurogenesis, starting from the cell fate transition of transcriptionally primed cortical progenitors. A particular emphasis will be placed on RBPs with mostly conserved but also divergent evolutionary functions in neural progenitors across different species. RBPs, when considered in the context of the fascinating process of neocortical development, deserve to be main protagonists in the story of the evolution and development of the neocortex.
Collapse
|
5
|
Franchini LF. Genetic Mechanisms Underlying Cortical Evolution in Mammals. Front Cell Dev Biol 2021; 9:591017. [PMID: 33659245 PMCID: PMC7917222 DOI: 10.3389/fcell.2021.591017] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/08/2021] [Indexed: 12/13/2022] Open
Abstract
The remarkable sensory, motor, and cognitive abilities of mammals mainly depend on the neocortex. Thus, the emergence of the six-layered neocortex in reptilian ancestors of mammals constitutes a fundamental evolutionary landmark. The mammalian cortex is a columnar epithelium of densely packed cells organized in layers where neurons are generated mainly in the subventricular zone in successive waves throughout development. Newborn cells move away from their site of neurogenesis through radial or tangential migration to reach their specific destination closer to the pial surface of the same or different cortical area. Interestingly, the genetic programs underlying neocortical development diversified in different mammalian lineages. In this work, I will review several recent studies that characterized how distinct transcriptional programs relate to the development and functional organization of the neocortex across diverse mammalian lineages. In some primates such as the anthropoids, the neocortex became extremely large, especially in humans where it comprises around 80% of the brain. It has been hypothesized that the massive expansion of the cortical surface and elaboration of its connections in the human lineage, has enabled our unique cognitive capacities including abstract thinking, long-term planning, verbal language and elaborated tool making capabilities. I will also analyze the lineage-specific genetic changes that could have led to the modification of key neurodevelopmental events, including regulation of cell number, neuronal migration, and differentiation into specific phenotypes, in order to shed light on the evolutionary mechanisms underlying the diversity of mammalian brains including the human brain.
Collapse
Affiliation(s)
- Lucía Florencia Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
6
|
Tomita H, Cornejo F, Aranda-Pino B, Woodard CL, Rioseco CC, Neel BG, Alvarez AR, Kaplan DR, Miller FD, Cancino GI. The Protein Tyrosine Phosphatase Receptor Delta Regulates Developmental Neurogenesis. Cell Rep 2021; 30:215-228.e5. [PMID: 31914388 DOI: 10.1016/j.celrep.2019.11.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 10/10/2019] [Accepted: 11/07/2019] [Indexed: 12/26/2022] Open
Abstract
PTPRD is a receptor protein tyrosine phosphatase that is genetically associated with neurodevelopmental disorders. Here, we asked whether Ptprd mutations cause aberrant neural development by perturbing neurogenesis in the murine cortex. We show that loss of Ptprd causes increases in neurogenic transit-amplifying intermediate progenitor cells and cortical neurons and perturbations in neuronal localization. These effects are intrinsic to neural precursor cells since acute Ptprd knockdown causes similar perturbations. PTPRD mediates these effects by dephosphorylating receptor tyrosine kinases, including TrkB and PDGFRβ, and loss of Ptprd causes the hyperactivation of TrkB and PDGFRβ and their downstream MEK-ERK signaling pathway in neural precursor cells. Moreover, inhibition of aberrant TrkB or MEK activation rescues the increased neurogenesis caused by knockdown or homozygous loss of Ptprd. These results suggest that PTPRD regulates receptor tyrosine kinases to ensure appropriate numbers of intermediate progenitor cells and neurons, suggesting a mechanism for its genetic association with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hideaki Tomita
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1X8, ON, Canada
| | - Francisca Cornejo
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - Begoña Aranda-Pino
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - Cameron L Woodard
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1X8, ON, Canada
| | - Constanza C Rioseco
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1X8, ON, Canada
| | - Benjamin G Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Alejandra R Alvarez
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - David R Kaplan
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1X8, ON, Canada; Institute of Medical Science, University of Toronto, Toronto M5S 1A8, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, ON, Canada
| | - Freda D Miller
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1X8, ON, Canada; Institute of Medical Science, University of Toronto, Toronto M5S 1A8, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, ON, Canada; Department of Physiology, University of Toronto, Toronto M5S 1A8, ON, Canada
| | - Gonzalo I Cancino
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto M5G 1X8, ON, Canada; Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile.
| |
Collapse
|
7
|
Abstract
The mammalian cerebral cortex is the pinnacle of brain evolution, reaching its maximum complexity in terms of neuron number, diversity and functional circuitry. The emergence of this outstanding complexity begins during embryonic development, when a limited number of neural stem and progenitor cells manage to generate myriads of neurons in the appropriate numbers, types and proportions, in a process called neurogenesis. Here we review the current knowledge on the regulation of cortical neurogenesis, beginning with a description of the types of progenitor cells and their lineage relationships. This is followed by a review of the determinants of neuron fate, the molecular and genetic regulatory mechanisms, and considerations on the evolution of cortical neurogenesis in vertebrates leading to humans. We finish with an overview on how dysregulation of neurogenesis is a leading cause of human brain malformations and functional disabilities.
Collapse
Affiliation(s)
- Ana Villalba
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Magdalena Götz
- Institute for Stem Cell Research, Helmholtz Zentrum München & Biomedical Center, Ludwig-Maximilians Universitaet, Planegg-Martinsried, Germany
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain.
| |
Collapse
|
8
|
García-Moreno F, Molnár Z. Variations of telencephalic development that paved the way for neocortical evolution. Prog Neurobiol 2020; 194:101865. [PMID: 32526253 PMCID: PMC7656292 DOI: 10.1016/j.pneurobio.2020.101865] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022]
Abstract
Charles Darwin stated, "community in embryonic structure reveals community of descent". Thus, to understand how the neocortex emerged during mammalian evolution we need to understand the evolution of the development of the pallium, the source of the neocortex. In this article, we review the variations in the development of the pallium that enabled the production of the six-layered neocortex. We propose that an accumulation of subtle modifications from very early brain development accounted for the diversification of vertebrate pallia and the origin of the neocortex. Initially, faint differences of expression of secretable morphogens promote a wide variety in the proportions and organization of sectors of the early pallium in different vertebrates. It prompted different sectors to host varied progenitors and distinct germinative zones. These cells and germinative compartments generate diverse neuronal populations that migrate and mix with each other through radial and tangential migrations in a taxon-specific fashion. Together, these early variations had a profound influence on neurogenetic gradients, lamination, positioning, and connectivity. Gene expression, hodology, and physiological properties of pallial neurons are important features to suggest homologies, but the origin of cells and their developmental trajectory are fundamental to understand evolutionary changes. Our review compares the development of the homologous pallial sectors in sauropsids and mammals, with a particular focus on cell lineage, in search of the key changes that led to the appearance of the mammalian neocortex.
Collapse
Affiliation(s)
- Fernando García-Moreno
- Achucarro Basque Center for Neuroscience, Scientific Park of the University of the Basque Country (UPV/EHU), 48940, Leioa, Spain; IKERBASQUE Foundation, María Díaz de Haro 3, 6th Floor, 48013, Bilbao, Spain; Department of Neuroscience, Faculty of Medicine and Odontology, UPV/EHU, Barrio Sarriena s/n, 48940, Leioa, Bizkaia, Spain.
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, OX1 3QX, UK.
| |
Collapse
|
9
|
Cárdenas A, Borrell V. Molecular and cellular evolution of corticogenesis in amniotes. Cell Mol Life Sci 2020; 77:1435-1460. [PMID: 31563997 PMCID: PMC11104948 DOI: 10.1007/s00018-019-03315-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/03/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
The cerebral cortex varies dramatically in size and complexity between amniotes due to differences in neuron number and composition. These differences emerge during embryonic development as a result of variations in neurogenesis, which are thought to recapitulate modifications occurred during evolution that culminated in the human neocortex. Here, we review work from the last few decades leading to our current understanding of the evolution of neurogenesis and size of the cerebral cortex. Focused on specific examples across vertebrate and amniote phylogeny, we discuss developmental mechanisms regulating the emergence, lineage, complexification and fate of cortical germinal layers and progenitor cell types. At the cellular level, we discuss the fundamental impact of basal progenitor cells and the advent of indirect neurogenesis on the increased number and diversity of cortical neurons and layers in mammals, and on cortex folding. Finally, we discuss recent work that unveils genetic and molecular mechanisms underlying this progressive expansion and increased complexity of the amniote cerebral cortex during evolution, with a particular focus on those leading to human-specific features. Whereas new genes important in human brain development emerged the recent hominid lineage, regulation of the patterns and levels of activity of highly conserved signaling pathways are beginning to emerge as mechanisms of central importance in the evolutionary increase in cortical size and complexity across amniotes.
Collapse
Affiliation(s)
- Adrián Cárdenas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernández, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernández, 03550, Sant Joan d'Alacant, Alicante, Spain.
| |
Collapse
|
10
|
Schmidt M. Evolution of the Hypothalamus and Inferior Lobe in Ray-Finned Fishes. BRAIN, BEHAVIOR AND EVOLUTION 2020; 95:302-316. [PMID: 32203953 DOI: 10.1159/000505898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/12/2020] [Indexed: 11/19/2022]
Abstract
The inferior lobes are prominent bilateral brain areas in the hypothalamus of neopterygians among the ray-finned fishes. They are known as multisensory integration centers. As such, they should play a major role in fish evolution. In this study, a comparative morphometric analysis was performed. The morphology of the hypothalamus, where the inferior lobe is considered as fully developed first in Lepisosteus, was then re-examined. One hundred brains from different species of 60 families of ray-finned fishes were stained with cresyl violet and embedded in methacrylate. They were then cut on a microtome while conducting block-face imaging. The volumes were determined for the whole brain, brain areas, and nuclei. Since visual input represents a major sensory input for the inferior lobe, the nucleus glomerulosus, a visual-related nucleus in paracanthopterygian and acanthopterygian teleosts, and the tectum opticum were included in the investigations. The morphometric analysis revealed that the relative volume of the inferior lobes increases significantly from species of the Lepisosteiformes to the Tetraodontiformes. In addition, a positive correlation was detected between the relative volume of the inferior lobes and either the relative volume of the nucleus glomerulosus or the relative volume of the tectum opticum. These correlations, in combination with findings from previous hodological and behavioral studies, give rise to the speculation that the inferior lobes may be involved in higher cognitive processes and complex social interactions.
Collapse
|
11
|
Yang H, Kim J, Kim Y, Jang SW, Sestan N, Shim S. Cux2 expression regulated by Lhx2 in the upper layer neurons of the developing cortex. Biochem Biophys Res Commun 2020; 521:874-879. [PMID: 31708105 DOI: 10.1016/j.bbrc.2019.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/01/2019] [Indexed: 12/20/2022]
Abstract
The laminar structure, a unique feature of the mammalian cerebrum, is formed by a number of genes in a highly complex process. The pyramidal neurons that make up each layer of the cerebrum are functionally characterized by specific gene expressions. In particular, Cux1 and Cux2, which are specifically expressed in layer II-IV neurons, are known to regulate dendritic branching, spine morphology, and synapse formation. However, it is still unknown how their expression is regulated transcriptionally. Here we constructed Cux2-mCherry transgenic mice that reproduce the cortical layer II-IV-specific expression of Cux2, a member of the Cut/Cux/CDP family, using BAC transgenesis and a variety of coordinated cortical layer markers that are known to date. Our immunohistochemistry analysis shows that mCherry was expressed in cortical layer II-IV and the corpus callosum in the same way as endogenous Cux2 without ectopic expression. We also identified a region of 220 bp that is highly conserved in mammals and controls specific cerebral expression of Cux2, using comparative genome analysis and in vivo reporter assays. Furthermore, we confirm that Lhx2, whose expression in cortical layer II-IV is similar to that of the Cux2 enhancer, can act as a transcriptional activator. These results suggest that cortical layer II-IV expression of Cux2 can be regulated by the interaction of Cux2-E1 and Lhx2, and that their failure to co-regulate is associated with neurodevelopmental disorders such as autism and schizophrenia.
Collapse
Affiliation(s)
- Hayoung Yang
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jiwoo Kim
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Yujin Kim
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Sung-Wuk Jang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, 06510, USA.
| | - Sungbo Shim
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
12
|
Polioudakis D, de la Torre-Ubieta L, Langerman J, Elkins AG, Shi X, Stein JL, Vuong CK, Nichterwitz S, Gevorgian M, Opland CK, Lu D, Connell W, Ruzzo EK, Lowe JK, Hadzic T, Hinz FI, Sabri S, Lowry WE, Gerstein MB, Plath K, Geschwind DH. A Single-Cell Transcriptomic Atlas of Human Neocortical Development during Mid-gestation. Neuron 2019; 103:785-801.e8. [PMID: 31303374 DOI: 10.1016/j.neuron.2019.06.011] [Citation(s) in RCA: 284] [Impact Index Per Article: 56.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/20/2019] [Accepted: 06/12/2019] [Indexed: 01/05/2023]
Abstract
We performed RNA sequencing on 40,000 cells to create a high-resolution single-cell gene expression atlas of developing human cortex, providing the first single-cell characterization of previously uncharacterized cell types, including human subplate neurons, comparisons with bulk tissue, and systematic analyses of technical factors. These data permit deconvolution of regulatory networks connecting regulatory elements and transcriptional drivers to single-cell gene expression programs, significantly extending our understanding of human neurogenesis, cortical evolution, and the cellular basis of neuropsychiatric disease. We tie cell-cycle progression with early cell fate decisions during neurogenesis, demonstrating that differentiation occurs on a transcriptomic continuum; rather than only expressing a few transcription factors that drive cell fates, differentiating cells express broad, mixed cell-type transcriptomes before telophase. By mapping neuropsychiatric disease genes to cell types, we implicate dysregulation of specific cell types in ASD, ID, and epilepsy. We developed CoDEx, an online portal to facilitate data access and browsing.
Collapse
Affiliation(s)
- Damon Polioudakis
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Luis de la Torre-Ubieta
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Justin Langerman
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Andrew G Elkins
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Xu Shi
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Jason L Stein
- Department of Genetics & UNC Neuroscience Center, University of North Carolina, Chapel Hill, Chapel Hill, NC, USA
| | - Celine K Vuong
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA, USA
| | - Susanne Nichterwitz
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Melinda Gevorgian
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Biology, CSUN, Northridge, CA, USA
| | - Carli K Opland
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Daning Lu
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - William Connell
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Elizabeth K Ruzzo
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Jennifer K Lowe
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Tarik Hadzic
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Flora I Hinz
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Shan Sabri
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - William E Lowry
- Department of Molecular, Cell and Developmental Biology, UCLA, Los Angeles, CA, USA
| | - Mark B Gerstein
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA; Department of Computer Science, Yale University, New Haven, CT 06520, USA; Department of Statistics and Data Science, Yale University, New Haven, CT 06520, USA
| | - Kathrin Plath
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA; Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Karzbrun E, Reiner O. Brain Organoids-A Bottom-Up Approach for Studying Human Neurodevelopment. Bioengineering (Basel) 2019; 6:E9. [PMID: 30669275 PMCID: PMC6466401 DOI: 10.3390/bioengineering6010009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/08/2019] [Accepted: 01/11/2019] [Indexed: 12/25/2022] Open
Abstract
Brain organoids have recently emerged as a three-dimensional tissue culture platform to study the principles of neurodevelopment and morphogenesis. Importantly, brain organoids can be derived from human stem cells, and thus offer a model system for early human brain development and human specific disorders. However, there are still major differences between the in vitro systems and in vivo development. This is in part due to the challenge of engineering a suitable culture platform that will support proper development. In this review, we discuss the similarities and differences of human brain organoid systems in comparison to embryonic development. We then describe how organoids are used to model neurodevelopmental diseases. Finally, we describe challenges in organoid systems and how to approach these challenges using complementary bioengineering techniques.
Collapse
Affiliation(s)
- Eyal Karzbrun
- Kavli Institute for Theoretical Physics and Department of Physics, University of California, Santa Barbara, CA 93106, USA.
| | - Orly Reiner
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
14
|
Kaas JH. The origin and evolution of neocortex: From early mammals to modern humans. PROGRESS IN BRAIN RESEARCH 2019; 250:61-81. [DOI: 10.1016/bs.pbr.2019.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
15
|
Zhang L, Mubarak T, Chen Y, Lee T, Pollock A, Sun T. Counter-Balance Between Gli3 and miR-7 Is Required for Proper Morphogenesis and Size Control of the Mouse Brain. Front Cell Neurosci 2018; 12:259. [PMID: 30210296 PMCID: PMC6121149 DOI: 10.3389/fncel.2018.00259] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 07/30/2018] [Indexed: 12/25/2022] Open
Abstract
Brain morphogenesis requires precise regulation of multiple genes to control specification of distinct neural progenitors (NPs) and neuronal production. Dysregulation of these genes results in severe brain malformation such as macrocephaly and microcephaly. Despite studies of the effect of individual pathogenic genes, the counter-balance between multiple factors in controlling brain size remains unclear. Here we show that cortical deletion of Gli3 results in enlarged brain and folding structures in the cortical midline at the postnatal stage, which is mainly caused by the increased percentage of intermediate progenitors (IPs) and newborn neurons. In addition, dysregulation of neuronal migration also contributes to the folding defects in the cortical midline region. Knockdown of microRNA (miRNA) miR-7 can rescue abnormal brain morphology in Gli3 knockout mice by recovering progenitor specification, neuronal production and migration through a counter-balance of the Gli3 activity. Moreover, miR-7 likely exerts its function through silencing target gene Pax6. Our results indicate that proper brain morphogenesis is an outcome of interactive regulations of multiple molecules such as Gli3 and miR-7. Because miRNAs are easy to synthesize and deliver, miR-7 could be a potential therapeutic means to macrocephaly caused by Gli3-deficiency.
Collapse
Affiliation(s)
- Longbin Zhang
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Taufif Mubarak
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Yase Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Trevor Lee
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Andrew Pollock
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| |
Collapse
|
16
|
Sauerland C, Menzies BR, Glatzle M, Seeger J, Renfree MB, Fietz SA. The Basal Radial Glia Occurs in Marsupials and Underlies the Evolution of an Expanded Neocortex in Therian Mammals. Cereb Cortex 2018; 28:145-157. [PMID: 29253253 DOI: 10.1093/cercor/bhw360] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/30/2016] [Indexed: 12/13/2022] Open
Abstract
A hallmark of mammalian brain evolution is the emergence of the neocortex, which has expanded in all mammalian infraclasses (Eutheria, Marsupialia, Monotremata). In eutherians, neocortical neurons derive from distinct neural stem and progenitor cells (NPCs). However, precise data on the presence and abundance of the NPCs, especially of basal radial glia (bRG), in the neocortex of marsupials are lacking. This study characterized and quantified the NPCs in the developing neocortex of a marsupial, the tammar wallaby (Macropus eugenii). Our data demonstrate that its neocortex is characterized by high NPC diversity. Importantly, we show that bRG exist at high relative abundance in the tammar indicating that this cell type is not specific to the eutherian neocortex and that similar mechanisms may underlie the formation of an expanded neocortex in eutherian and marsupial mammals. We also show that bRG are likely to have been present in the therian ancestor, so did not emerge independently in the eutherian and marsupial lineages. Moreover, our data support the concept that changes in multiple parameters contribute to neocortex expansion and demonstrate the importance of bRG and other NPCs for the development and expansion of the mammalian neocortex.
Collapse
Affiliation(s)
- Christine Sauerland
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| | - Brandon R Menzies
- School of BioSciences, The University of Melbourne, Victoria 3010, Melbourne, Australia
| | - Megan Glatzle
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| | - Johannes Seeger
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| | - Marilyn B Renfree
- School of BioSciences, The University of Melbourne, Victoria 3010, Melbourne, Australia
| | - Simone A Fietz
- Institute of Veterinary Anatomy, Histology and Embryology, University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
17
|
Zhang H, Zhang L, Sun T. Cohesive Regulation of Neural Progenitor Development by microRNA miR-26, Its Host Gene Ctdsp and Target Gene Emx2 in the Mouse Embryonic Cerebral Cortex. Front Mol Neurosci 2018. [PMID: 29515367 PMCID: PMC5825903 DOI: 10.3389/fnmol.2018.00044] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Proper proliferation and differentiation of neural progenitors (NPs) in the developing cerebral cortex are critical for normal brain formation and function. Emerging evidence has shown the importance of microRNAs (miRNAs) in regulating cortical development and the etiology of neurological disorders. Here we show that miR-26 is co-expressed with its host gene Ctdsp in the mouse embryonic cortex. We demonstrate that similar to its host gene Ctdsp2, miR-26 positively regulates proliferation of NPs through controlling the cell-cycle progression, by using miR-26 overexpression and sponge approaches. On the contrary, miR-26 target gene Emx2 limits expansion of cortical NPs, and promotes transcription of miR-26 host gene Ctdsp. Our study suggests that miR-26, its target Emx2 and its host gene Ctdsp cohesively regulate proliferation of NPs during the mouse cortical development.
Collapse
Affiliation(s)
- Haijun Zhang
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York, NY, United States.,Department of Genetic Medicine, Weill Cornell Medical College, Cornell University, New York, NY, United States
| | - Longbin Zhang
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Tao Sun
- Department of Cell and Developmental Biology, Weill Cornell Medical College, Cornell University, New York, NY, United States.,Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| |
Collapse
|
18
|
Furlan G, Cuccioli V, Vuillemin N, Dirian L, Muntasell AJ, Coolen M, Dray N, Bedu S, Houart C, Beaurepaire E, Foucher I, Bally-Cuif L. Life-Long Neurogenic Activity of Individual Neural Stem Cells and Continuous Growth Establish an Outside-In Architecture in the Teleost Pallium. Curr Biol 2017; 27:3288-3301.e3. [PMID: 29107546 PMCID: PMC5678050 DOI: 10.1016/j.cub.2017.09.052] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/14/2017] [Accepted: 09/25/2017] [Indexed: 01/08/2023]
Abstract
Spatiotemporal variations of neurogenesis are thought to account for the evolution of brain shape. In the dorsal telencephalon (pallium) of vertebrates, it remains unresolved which ancestral neurogenesis mode prefigures the highly divergent cytoarchitectures that are seen in extant species. To gain insight into this question, we developed genetic tools to generate here the first 4-dimensional (3D + birthdating time) map of pallium construction in the adult teleost zebrafish. Using a Tet-On-based genetic birthdating strategy, we identify a “sequential stacking” construction mode where neurons derived from the zebrafish pallial germinal zone arrange in outside-in, age-related layers from a central core generated during embryogenesis. We obtained no evidence for overt radial or tangential neuronal migrations. Cre-lox-mediated tracing, which included following Brainbow clones, further demonstrates that this process is sustained by the persistent neurogenic activity of individual pallial neural stem cells (NSCs) from embryo to adult. Together, these data demonstrate that the spatiotemporal control of NSC activity is an important driver of the macroarchitecture of the zebrafish adult pallium. This simple mode of pallium construction shares distinct traits with pallial genesis in mammals and non-mammalian amniotes such as birds or reptiles, suggesting that it may exemplify the basal layout from which vertebrate pallial architectures were elaborated. Neurons of the teleost pallium are arranged in concentric age-dependent layers Neurons of the central pallial domain, Dc, are born during embryogenesis Most pallial neurons are generated from ventricular her4-positive radial glia The majority of individual pallial radial glia are neurogenic throughout life
Collapse
Affiliation(s)
- Giacomo Furlan
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Valentina Cuccioli
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Nelly Vuillemin
- Laboratory for Optics and Biosciences, École Polytechnique, CNRS UMR 7645 and INSERM U1182, 91128 Palaiseau, France
| | - Lara Dirian
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France
| | - Anna Janue Muntasell
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London SE1 1UL, UK
| | - Marion Coolen
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Nicolas Dray
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Sébastien Bedu
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Corinne Houart
- Centre for Developmental Neurobiology and MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London SE1 1UL, UK
| | - Emmanuel Beaurepaire
- Laboratory for Optics and Biosciences, École Polytechnique, CNRS UMR 7645 and INSERM U1182, 91128 Palaiseau, France
| | - Isabelle Foucher
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France.
| | - Laure Bally-Cuif
- Team Zebrafish Neurogenetics, Paris-Saclay Institute for Neuroscience (Neuro-PSI), UMR 9197, CNRS-Université Paris-Sud, Avenue de la Terrasse, 91190 Gif-sur-Yvette, France; Unit Zebrafish Neurogenetics, Developmental and Stem Cell Biology Department, Institut Pasteur, 25 Rue du Dr Roux, 75015 Paris, France; CNRS UMR 3738, 25 Rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
19
|
Dynamic behaviour of human neuroepithelial cells in the developing forebrain. Nat Commun 2017; 8:14167. [PMID: 28139695 PMCID: PMC5290330 DOI: 10.1038/ncomms14167] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 12/05/2016] [Indexed: 11/09/2022] Open
Abstract
To understand how diverse progenitor cells contribute to human neocortex development, we examined forebrain progenitor behaviour using timelapse imaging. Here we find that cell cycle dynamics of human neuroepithelial (NE) cells differ from radial glial (RG) cells in both primary tissue and in stem cell-derived organoids. NE cells undergoing proliferative, symmetric divisions retract their basal processes, and both daughter cells regrow a new process following cytokinesis. The mitotic retraction of the basal process is recapitulated by NE cells in cerebral organoids generated from human-induced pluripotent stem cells. In contrast, RG cells undergoing vertical cleavage retain their basal fibres throughout mitosis, both in primary tissue and in older organoids. Our findings highlight developmentally regulated changes in mitotic behaviour that may relate to the role of RG cells to provide a stable scaffold for neuronal migration, and suggest that the transition in mitotic dynamics can be studied in organoid models. The dynamics of progenitor cells in human neocortex development has not been studied directly. Here, the authors timelapse image human neuroepithelial (NE) and radial glial (RG) cells in embryonic brain slices and find properties of NE cells and RG that are mimicked in cerebral organoids.
Collapse
|
20
|
Carson R, Monaghan-Nichols AP, DeFranco DB, Rudine AC. Effects of antenatal glucocorticoids on the developing brain. Steroids 2016; 114:25-32. [PMID: 27343976 PMCID: PMC5052110 DOI: 10.1016/j.steroids.2016.05.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 05/25/2016] [Accepted: 05/30/2016] [Indexed: 01/13/2023]
Abstract
Glucocorticoids (GCs) regulate distinct physiological processes in the developing fetus, in particular accelerating organ maturation that enables the fetus to survive outside the womb. In preterm birth, the developing fetus does not receive sufficient exposure to endogenous GCs in utero for proper organ development predisposing the neonate to complications including intraventricular hemorrhage, respiratory distress syndrome (RDS) and necrotizing enterocolitis (NEC). Synthetic GCs (sGCs) have proven useful in the prevention of these complications since they are able to promote the rapid maturation of underdeveloped organs present in the fetus. While these drugs have proven to be clinically effective in the prevention of IVH, RDS and NEC, they may also trigger adverse developmental side effects. This review will examine the current clinical use of antenatal sGC therapy in preterm birth, their placental metabolism, and their effects on the developing brain.
Collapse
Affiliation(s)
- Ross Carson
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - A Paula Monaghan-Nichols
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Neurobiology, United States
| | - Donald B DeFranco
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Pharmacology and Chemical Biology, United States
| | - Anthony C Rudine
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Department of Pediatrics, Division of Newborn Medicine, United States.
| |
Collapse
|
21
|
Amamoto R, Huerta VGL, Takahashi E, Dai G, Grant AK, Fu Z, Arlotta P. Adult axolotls can regenerate original neuronal diversity in response to brain injury. eLife 2016; 5. [PMID: 27156560 PMCID: PMC4861602 DOI: 10.7554/elife.13998] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/07/2016] [Indexed: 12/11/2022] Open
Abstract
The axolotl can regenerate multiple organs, including the brain. It remains, however, unclear whether neuronal diversity, intricate tissue architecture, and axonal connectivity can be regenerated; yet, this is critical for recovery of function and a central aim of cell replacement strategies in the mammalian central nervous system. Here, we demonstrate that, upon mechanical injury to the adult pallium, axolotls can regenerate several of the populations of neurons present before injury. Notably, regenerated neurons acquire functional electrophysiological traits and respond appropriately to afferent inputs. Despite the ability to regenerate specific, molecularly-defined neuronal subtypes, we also uncovered previously unappreciated limitations by showing that newborn neurons organize within altered tissue architecture and fail to re-establish the long-distance axonal tracts and circuit physiology present before injury. The data provide a direct demonstration that diverse, electrophysiologically functional neurons can be regenerated in axolotls, but challenge prior assumptions of functional brain repair in regenerative species. DOI:http://dx.doi.org/10.7554/eLife.13998.001 Humans and other mammals have a very limited ability to regenerate new neurons in the brain to replace those that have been injured or damaged. In striking contrast, some animals like fish and salamanders are capable of filling in injured brain regions with new neurons. This is a complex task, as the brain is composed of many different types of neurons that are connected to each other in a highly organized manner across both short and long distances. The extent to which even the most regenerative species can build new brain regions was not known. Understanding any limitations will help to set realistic expectations for the success of potential treatments that aim to replace neurons in mammals. Amamoto et al. found that the brain of the axolotl, a species of salamander, could selectively regenerate the specific types of neurons that were damaged. This finding suggests that the brain is able to somehow sense which types of neurons are injured. The new neurons were able to mature into functional neurons, but they were limited in their ability to reconnect to their original, distant target neurons. More research is now needed to investigate how the axolotl brain recognizes which types of neurons have been damaged. It will also be important to understand which cells respond to the injury to give rise to the new neurons that fill the injury site, and to uncover the molecules that are important for governing this regenerative process. DOI:http://dx.doi.org/10.7554/eLife.13998.002
Collapse
Affiliation(s)
- Ryoji Amamoto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | | | - Emi Takahashi
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Guangping Dai
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Aaron K Grant
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States
| | - Zhanyan Fu
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, United States
| |
Collapse
|
22
|
Montiel JF, Vasistha NA, Garcia-Moreno F, Molnár Z. From sauropsids to mammals and back: New approaches to comparative cortical development. J Comp Neurol 2016; 524:630-45. [PMID: 26234252 PMCID: PMC4832283 DOI: 10.1002/cne.23871] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 07/21/2015] [Accepted: 07/21/2015] [Indexed: 01/31/2023]
Abstract
Evolution of the mammalian neocortex (isocortex) has been a persisting problem in neurobiology. While recent studies have attempted to understand the evolutionary expansion of the human neocortex from rodents, similar approaches have been used to study the changes between reptiles, birds, and mammals. We review here findings from the past decades on the development, organization, and gene expression patterns in various extant species. This review aims to compare cortical cell numbers and neuronal cell types to the elaboration of progenitor populations and their proliferation in these species. Several progenitors, such as the ventricular radial glia, the subventricular intermediate progenitors, and the subventricular (outer) radial glia, have been identified but the contribution of each to cortical layers and cell types through specific lineages, their possible roles in determining brain size or cortical folding, are not yet understood. Across species, larger, more diverse progenitors relate to cortical size and cell diversity. The challenge is to relate the radial and tangential expansion of the neocortex to the changes in the proliferative compartments during mammalian evolution and with the changes in gene expression and lineages evident in various sectors of the developing brain. We also review the use of recent lineage tracing and transcriptomic approaches to revisit theories and to provide novel understanding of molecular processes involved in specification of cortical regions.
Collapse
Affiliation(s)
- Juan F Montiel
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Centre for Biomedical Research, Facultad de Medicina, Universidad Diego Portales, Santiago, Chile
| | - Navneet A Vasistha
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Scotland, UK
| | | | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Yang N, Liu H, Jiang Y, Zheng J, Li DM, Ji C, Liu YY, Zuo PP. Lactulose enhances neuroplasticity to improve cognitive function in early hepatic encephalopathy. Neural Regen Res 2015; 10:1457-62. [PMID: 26604907 PMCID: PMC4625512 DOI: 10.4103/1673-5374.165516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lactulose is known to improve cognitive function in patients with early hepatic encephalopathy; however, the underlying mechanism remains poorly understood. In the present study, we investigated the behavioral and neurochemical effects of lactulose in a rat model of early hepatic encephalopathy induced by carbon tetrachloride. Immunohistochemistry showed that lactulose treatment promoted neurogenesis and increased the number of neurons and astrocytes in the hippocampus. Moreover, lactulose-treated rats showed shorter escape latencies than model rats in the Morris water maze, indicating that lactulose improved the cognitive impairments caused by hepatic encephalopathy. The present findings suggest that lactulose effectively improves cognitive function by enhancing neuroplasticity in a rat model of early hepatic encephalopathy.
Collapse
Affiliation(s)
- Nan Yang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - He Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yao Jiang
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Ji Zheng
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Dong-Mei Li
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Chao Ji
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yan-Yong Liu
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Ping-Ping Zuo
- Department of Pharmacology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
CHCHD2 is down-regulated in neuronal cells differentiated from iPS cells derived from patients with lissencephaly. Genomics 2015; 106:196-203. [DOI: 10.1016/j.ygeno.2015.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/17/2015] [Accepted: 07/01/2015] [Indexed: 12/20/2022]
|
25
|
Brunjes PC, Osterberg SK. Developmental Markers Expressed in Neocortical Layers Are Differentially Exhibited in Olfactory Cortex. PLoS One 2015; 10:e0138541. [PMID: 26407299 PMCID: PMC4583488 DOI: 10.1371/journal.pone.0138541] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 08/31/2015] [Indexed: 11/28/2022] Open
Abstract
Neurons in the cerebral cortex stratify on the basis of their time of origin, axonal terminations and the molecular identities assigned during early development. Olfactory cortices share many feature with the neocortex, including clear lamination and similar cell types. The present study demonstrates that the markers differentially expressed in the projection neurons of the cerebral cortex are also found in olfactory areas. Three of the four regions examined (pars principalis of the anterior olfactory nucleus: AONpP, anterior and posterior piriform cortices: APC, PPC, and the olfactory tubercle) expressed transcription factors found in deep or superficial neurons in the developing neocortex, though large differences were found between areas. For example, while the AONpP, APC and PPC all broadly expressed the deep cortical marker CTIP2, NOR1 (NR4a3) levels were higher in AONpP and DAARP-32 was more prevalent in the APC and PPC. Similar findings were encountered for superficial cortical markers: all three regions broadly expressed CUX1, but CART was only observed in the APC and PPC. Furthermore, regional variations were observed even within single structures (e.g., NOR1 was found primarily in in the dorsal region of AONpP and CART expression was observed in a discrete band in the middle of layer 2 of both the APC and PPC). Experiments using the mitotic marker EDU verified that the olfactory cortices and neocortex share similar patterns of neuronal production: olfactory cells that express markers found in the deep neocortex are produced earlier than those that express superficial makers. Projection neurons were filled by retrograde tracers injected into the olfactory bulb to see if olfactory neurons with deep and superficial markers had different axonal targets. Unlike the cerebral cortex, no specificity was observed: neurons with each of the transcription factors examined were found to be labelled. Together the results indicate that olfactory cortices are complex: they differ from each other and each is formed from a variable mosaic of neurons. The results suggest that the olfactory cortices are not merely a remnant architype of the primordial forebrain but varied and independent regions.
Collapse
Affiliation(s)
- Peter C. Brunjes
- Department Psychology, University of Virginia, Charlottesville, Virginia, 22904, United States of America
- * E-mail:
| | - Stephen K. Osterberg
- Department Psychology, University of Virginia, Charlottesville, Virginia, 22904, United States of America
| |
Collapse
|
26
|
Wong FK, Fei JF, Mora-Bermúdez F, Taverna E, Haffner C, Fu J, Anastassiadis K, Stewart AF, Huttner WB. Sustained Pax6 Expression Generates Primate-like Basal Radial Glia in Developing Mouse Neocortex. PLoS Biol 2015; 13:e1002217. [PMID: 26252244 PMCID: PMC4529158 DOI: 10.1371/journal.pbio.1002217] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 06/30/2015] [Indexed: 11/21/2022] Open
Abstract
The evolutionary expansion of the neocortex in mammals has been linked to enlargement of the subventricular zone (SVZ) and increased proliferative capacity of basal progenitors (BPs), notably basal radial glia (bRG). The transcription factor Pax6 is known to be highly expressed in primate, but not mouse, BPs. Here, we demonstrate that sustaining Pax6 expression selectively in BP-genic apical radial glia (aRG) and their BP progeny of embryonic mouse neocortex suffices to induce primate-like progenitor behaviour. Specifically, we conditionally expressed Pax6 by in utero electroporation using a novel, Tis21–CreERT2 mouse line. This expression altered aRG cleavage plane orientation to promote bRG generation, increased cell-cycle re-entry of BPs, and ultimately increased upper-layer neuron production. Upper-layer neuron production was also increased in double-transgenic mouse embryos with sustained Pax6 expression in the neurogenic lineage. Strikingly, increased BPs existed not only in the SVZ but also in the intermediate zone of the neocortex of these double-transgenic mouse embryos. In mutant mouse embryos lacking functional Pax6, the proportion of bRG among BPs was reduced. Our data identify specific Pax6 effects in BPs and imply that sustaining this Pax6 function in BPs could be a key aspect of SVZ enlargement and, consequently, the evolutionary expansion of the neocortex. "Humanizing" the expression of the transcription factor Pax6 in cortical progenitors in the developing mouse brain is sufficient to endow these progenitors with a primate-like proliferative capacity. During development, neural progenitors generate all cells that make up the mammalian brain. Differences in brain size among the various mammalian species are attributed to differences in the abundance and proliferative capacity of a specific class of neural progenitors called basal progenitors. Among these, a specific progenitor type called basal radial glia is thought to have played an important role during evolution in the expansion of the neocortex, the part of the brain associated with higher cognitive functions like conscious thought and language. In the neocortex, the expression of the transcription factor Pax6 in basal progenitors is low in rodents, but high in primates, including humans. In this study, we aimed to mimic the elevated expression pattern of Pax6 seen in humans in basal progenitors of the embryonic mouse neocortex. To this end, we generated a novel, transgenic mouse line that allows sustained expression of the Pax6 gene in basal progenitors. This elevated expression resulted in an increase in the generation of basal radial glia, in the proliferative capacity of basal progenitors, and, ultimately, in the number of neurons produced. Our findings demonstrate that altering the expression of a single transcription factor from a mouse to a human-like pattern suffices to induce a primate-like proliferative behaviour in neural progenitors, which is thought to underlie the evolutionary expansion of the neocortex.
Collapse
Affiliation(s)
- Fong Kuan Wong
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ji-Feng Fei
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Elena Taverna
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jun Fu
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | | | - A. Francis Stewart
- Biotechnology Center of the Technische Universität Dresden, Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail:
| |
Collapse
|
27
|
De Juan Romero C, Borrell V. Coevolution of radial glial cells and the cerebral cortex. Glia 2015; 63:1303-19. [PMID: 25808466 PMCID: PMC5008138 DOI: 10.1002/glia.22827] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/09/2015] [Indexed: 12/14/2022]
Abstract
Radial glia cells play fundamental roles in the development of the cerebral cortex, acting both as the primary stem and progenitor cells, as well as the guides for neuronal migration and lamination. These critical functions of radial glia cells in cortical development have been discovered mostly during the last 15 years and, more recently, seminal studies have demonstrated the existence of a remarkable diversity of additional cortical progenitor cell types, including a variety of basal radial glia cells with key roles in cortical expansion and folding, both in ontogeny and phylogeny. In this review, we summarize the main cellular and molecular mechanisms known to be involved in cerebral cortex development in mouse, as the currently preferred animal model, and then compare these with known mechanisms in other vertebrates, both mammal and nonmammal, including human. This allows us to present a global picture of how radial glia cells and the cerebral cortex seem to have coevolved, from reptiles to primates, leading to the remarkable diversity of vertebrate cortical phenotypes.
Collapse
Affiliation(s)
- Camino De Juan Romero
- Instituto De Neurociencias, Consejo Superior De Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan D'alacant, Spain
| | - Víctor Borrell
- Instituto De Neurociencias, Consejo Superior De Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan D'alacant, Spain
| |
Collapse
|
28
|
Hevner RF. Evolution of the mammalian dentate gyrus. J Comp Neurol 2015; 524:578-94. [PMID: 26179319 DOI: 10.1002/cne.23851] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/02/2015] [Accepted: 07/06/2015] [Indexed: 01/08/2023]
Abstract
The dentate gyrus (DG), a part of the hippocampal formation, has important functions in learning, memory, and adult neurogenesis. Compared with homologous areas in sauropsids (birds and reptiles), the mammalian DG is larger and exhibits qualitatively different phenotypes: 1) folded (C- or V-shaped) granule neuron layer, concave toward the hilus and delimited by a hippocampal fissure; 2) nonperiventricular adult neurogenesis; and 3) prolonged ontogeny, involving extensive abventricular (basal) migration and proliferation of neural stem and progenitor cells (NSPCs). Although gaps remain, available data indicate that these DG traits are present in all orders of mammals, including monotremes and marsupials. The exception is Cetacea (whales, dolphins, and porpoises), in which DG size, convolution, and adult neurogenesis have undergone evolutionary regression. Parsimony suggests that increased growth and convolution of the DG arose in stem mammals concurrently with nonperiventricular adult hippocampal neurogenesis and basal migration of NSPCs during development. These traits could all result from an evolutionary change that enhanced radial migration of NSPCs out of the periventricular zones, possibly by epithelial-mesenchymal transition, to colonize and maintain nonperiventricular proliferative niches. In turn, increased NSPC migration and clonal expansion might be a consequence of growth in the cortical hem (medial patterning center), which produces morphogens such as Wnt3a, generates Cajal-Retzius neurons, and is regulated by Lhx2. Finally, correlations between DG convolution and neocortical gyrification (or capacity for gyrification) suggest that enhanced abventricular migration and proliferation of NSPCs played a transformative role in growth and folding of neocortex as well as archicortex.
Collapse
Affiliation(s)
- Robert F Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, 98101
- Department of Neurological Surgery, University of Washington, Seattle, Washington, 98104
| |
Collapse
|
29
|
Neurodevelopmental LincRNA Microsyteny Conservation and Mammalian Brain Size Evolution. PLoS One 2015; 10:e0131818. [PMID: 26134977 PMCID: PMC4489927 DOI: 10.1371/journal.pone.0131818] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 06/07/2015] [Indexed: 11/19/2022] Open
Abstract
The mammalian neocortex has undergone repeated selection for increases and decreases in size and complexity, often over relatively short evolutionary time. But because probing developmental mechanisms across many species is experimentally unfeasible, it is unknown whether convergent morphologies in distantly related species are regulated by conserved developmental programs. In this work, we have taken advantage of the abundance of available mammalian genomes to find evidence of selection on genomic regions putatively regulating neurogenesis in large- versus small-brained species. Using published fetal human RNA-seq data, we show that the gene-neighborhood (i.e., microsynteny) of long intergenic non-coding RNAs (lincRNAs) implicated in cortical development is differentially conserved in large-brained species, lending support to the hypothesis that lincRNAs regulating neurogenesis are selectively lost in small-brained species. We provide evidence that this is not a phenomenon attributable to lincRNA expressed in all tissue types and is therefore likely to represent an adaptive function in the evolution of neurogenesis. A strong correlation between transcription factor-adjacency and lincRNA sequence conservation reinforces this conclusion.
Collapse
|
30
|
Bestman JE, Huang LC, Lee-Osbourne J, Cheung P, Cline HT. An in vivo screen to identify candidate neurogenic genes in the developing Xenopus visual system. Dev Biol 2015; 408:269-91. [PMID: 25818835 PMCID: PMC4584193 DOI: 10.1016/j.ydbio.2015.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 01/30/2015] [Accepted: 03/17/2015] [Indexed: 11/26/2022]
Abstract
Neurogenesis in the brain of Xenopus laevis continues throughout larval stages of development. We developed a 2-tier screen to identify candidate genes controlling neurogenesis in Xenopus optic tectum in vivo. First, microarray and NanoString analyses were used to identify candidate genes that were differentially expressed in Sox2-expressing neural progenitor cells or their neuronal progeny. Then an in vivo, time-lapse imaging-based screen was used to test whether morpholinos against 34 candidate genes altered neural progenitor cell proliferation or neuronal differentiation over 3 days in the optic tectum of intact Xenopus tadpoles. We co-electroporated antisense morpholino oligonucleotides against each of the candidate genes with a plasmid that drives GFP expression in Sox2-expressing neural progenitor cells and quantified the effects of morpholinos on neurogenesis. Of the 34 morpholinos tested, 24 altered neural progenitor cell proliferation or neuronal differentiation. The candidates which were tagged as differentially expressed and validated by the in vivo imaging screen include: actn1, arl9, eif3a, elk4, ephb1, fmr1-a, fxr1-1, fbxw7, fgf2, gstp1, hat1, hspa5, lsm6, mecp2, mmp9, and prkaca. Several of these candidates, including fgf2 and elk4, have known or proposed neurogenic functions, thereby validating our strategy to identify candidates. Genes with no previously demonstrated neurogenic functions, gstp1, hspa5 and lsm6, were identified from the morpholino experiments, suggesting that our screen successfully revealed unknown candidates. Genes that are associated with human disease, such as such as mecp2 and fmr1-a, were identified by our screen, providing the groundwork for using Xenopus as an experimental system to probe conserved disease mechanisms. Together the data identify candidate neurogenic regulatory genes and demonstrate that Xenopus is an effective experimental animal to identify and characterize genes that regulate neural progenitor cell proliferation and differentiation in vivo.
Collapse
Affiliation(s)
- Jennifer E Bestman
- Drug Discovery & Biomedical Sciences, The Medical University of South Carolina, Charleston, SC 29425, United States
| | - Lin-Chien Huang
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, United States
| | - Jane Lee-Osbourne
- University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Phillip Cheung
- Dart Neuroscience, LLC, San Diego, CA 92064, United States
| | - Hollis T Cline
- The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, United States.
| |
Collapse
|
31
|
Butts T, Green MJ, Wingate RJT. Development of the cerebellum: simple steps to make a 'little brain'. Development 2014; 141:4031-41. [PMID: 25336734 DOI: 10.1242/dev.106559] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cerebellum is a pre-eminent model for the study of neurogenesis and circuit assembly. Increasing interest in the cerebellum as a participant in higher cognitive processes and as a locus for a range of disorders and diseases make this simple yet elusive structure an important model in a number of fields. In recent years, our understanding of some of the more familiar aspects of cerebellar growth, such as its territorial allocation and the origin of its various cell types, has undergone major recalibration. Furthermore, owing to its stereotyped circuitry across a range of species, insights from a variety of species have contributed to an increasingly rich picture of how this system develops. Here, we review these recent advances and explore three distinct aspects of cerebellar development - allocation of the cerebellar anlage, the significance of transit amplification and the generation of neuronal diversity - each defined by distinct regulatory mechanisms and each with special significance for health and disease.
Collapse
Affiliation(s)
- Thomas Butts
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK School of Biological and Chemical Sciences, Queen Mary, University of London, London E1 4NS, UK
| | - Mary J Green
- National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Richard J T Wingate
- MRC Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| |
Collapse
|
32
|
Lewitus E, Kelava I, Kalinka AT, Tomancak P, Huttner WB. An adaptive threshold in mammalian neocortical evolution. PLoS Biol 2014; 12:e1002000. [PMID: 25405475 PMCID: PMC4236020 DOI: 10.1371/journal.pbio.1002000] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 10/09/2014] [Indexed: 01/19/2023] Open
Abstract
A study of the evolutionary history of cortical folding in mammals, its relationship to physiological and life-history traits and the underlying cortical progenitor behavior during embryogenesis, explains the diversity of folding we see across modern mammals. The diversity of neocortical folding among mammals can be explained by two distinct neurogenic programs, which give rise to mammals with a highly folded neocortex and mammals with slightly folded or unfolded neocortex, each occupying a distinct ecological niche. Expansion of the neocortex is a hallmark of human evolution. However, determining which adaptive mechanisms facilitated its expansion remains an open question. Here we show, using the gyrencephaly index (GI) and other physiological and life-history data for 102 mammalian species, that gyrencephaly is an ancestral mammalian trait. We find that variation in GI does not evolve linearly across species, but that mammals constitute two principal groups above and below a GI threshold value of 1.5, approximately equal to 109 neurons, which may be characterized by distinct constellations of physiological and life-history traits. By integrating data on neurogenic period, neuroepithelial founder pool size, cell-cycle length, progenitor-type abundances, and cortical neuron number into discrete mathematical models, we identify symmetric proliferative divisions of basal progenitors in the subventricular zone of the developing neocortex as evolutionarily necessary for generating a 14-fold increase in daily prenatal neuron production, traversal of the GI threshold, and thus establishment of two principal groups. We conclude that, despite considerable neuroanatomical differences, changes in the length of the neurogenic period alone, rather than any novel neurogenic progenitor lineage, are sufficient to explain differences in neuron number and neocortical size between species within the same principal group. What are the key differences in the development and evolution of the cerebral cortex that underlie the differences in its size and degree of folding across mammals? Here, we present phylogenetic evidence that the Jurassic era mammalian ancestor may have been a relatively large-brained species with a folded neocortex. We then show that variation in the degree of cortical folding (gyrencephaly index [GI]) does not evolve linearly across species, as previously assumed, but that mammals fall into two principal groups associated with distinct ecological niches: low-GI mammals (such as mice and tarsiers) and high-GI mammals (such as dolphins and humans), which are found to generate on average 14-fold more brain weight per day of gestation. This greater daily brain weight production in mammals with a highly folded neocortex requires a specific class of progenitor cell-type to adopt a special mode of cell division, which is absent in mammals with slightly folded or unfolded neocortices. Differences among mammals within the same GI group (high or low) are not due to different programming, but rather the result of differences in the length of the neurogenic period. So, the impressively large and folded human neocortex, which is three times the size of the chimpanzee neocortex, can be explained by a modest evolutionary extension of the neurogenic period with respect to its closest primate ancestors.
Collapse
Affiliation(s)
- Eric Lewitus
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (EL); (WBH)
| | - Iva Kelava
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Alex T. Kalinka
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Pavel Tomancak
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- * E-mail: (EL); (WBH)
| |
Collapse
|
33
|
Taverna E, Götz M, Huttner WB. The cell biology of neurogenesis: toward an understanding of the development and evolution of the neocortex. Annu Rev Cell Dev Biol 2014; 30:465-502. [PMID: 25000993 DOI: 10.1146/annurev-cellbio-101011-155801] [Citation(s) in RCA: 518] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neural stem and progenitor cells have a central role in the development and evolution of the mammalian neocortex. In this review, we first provide a set of criteria to classify the various types of cortical stem and progenitor cells. We then discuss the issue of cell polarity, as well as specific subcellular features of these cells that are relevant for their modes of division and daughter cell fate. In addition, cortical stem and progenitor cell behavior is placed into a tissue context, with consideration of extracellular signals and cell-cell interactions. Finally, the differences across species regarding cortical stem and progenitor cells are dissected to gain insight into key developmental and evolutionary mechanisms underlying neocortex expansion.
Collapse
Affiliation(s)
- Elena Taverna
- Max-Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany;
| | | | | |
Collapse
|
34
|
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: 53] [Impact Index Per Article: 5.3] [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
|
35
|
Charvet CJ. Distinct developmental growth patterns account for the disproportionate expansion of the rostral and caudal isocortex in evolution. Front Hum Neurosci 2014; 8:190. [PMID: 24782736 PMCID: PMC3986531 DOI: 10.3389/fnhum.2014.00190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 03/14/2014] [Indexed: 11/13/2022] Open
Abstract
In adulthood, the isocortex of several species is characterized by a gradient in neurons per unit of cortical surface area with fewer neurons per unit of cortical surface area in the rostral pole relative to the caudal pole. A gradient in neurogenesis timing predicts differences in neurons across the isocortex: neurons per unit of cortical surface area are fewer rostrally, where neurogenesis duration is short, and higher caudally where neurogenesis duration is longer. How species differences in neurogenesis duration impact cortical progenitor cells across its axis is not known. I estimated progenitor cells per unit of ventricular area across the rostro-caudal axis of the isocortex in cats (Felis catus) and in dogs (Canis familiaris) mostly before layers VI-II neurons are generated. I also estimated the ventricular length across the rostro-caudal axis at various stages of development in both species. These two species were chosen because neurogenesis duration in dogs is extended compared with cats. Caudally, cortical progenitors expand more tangentially and in numbers in dogs compared with cats. Rostrally, the cortical proliferative zone expands more tangentially in dogs compared with cats. However, the tangential expansion in the rostral cortical proliferative zone occurs without a concomitant increase in progenitor cell numbers. The tangential expansion of the ventricular surface in the rostral cortex is mediated by a reduction in cell density. These different developmental growth patterns account for the disproportionate expansion of the rostral (i.e., frontal cortex) and caudal cortex (e.g., primary visual cortex) when neurogenesis duration lengthens in evolution.
Collapse
|
36
|
Diverse behaviors of outer radial glia in developing ferret and human cortex. J Neurosci 2014; 34:2559-70. [PMID: 24523546 DOI: 10.1523/jneurosci.2645-13.2014] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The dramatic increase in neocortical size and folding during mammalian brain evolution has been attributed to the elaboration of the subventricular zone (SVZ) and the associated increase in neural progenitors. However, recent studies have shown that SVZ size and the abundance of resident progenitors do not directly predict cortical topography, suggesting that complex behaviors of the progenitors themselves may contribute to the overall size and shape of the adult cortex. Using time-lapse imaging, we examined the dynamic behaviors of SVZ progenitors in the ferret, a gyrencephalic carnivore, focusing our analysis on outer radial glial cells (oRGs). We identified a substantial population of oRGs by marker expression and their unique mode of division, termed mitotic somal translocation (MST). Ferret oRGs exhibited diverse behaviors in terms of division location, cleavage angle, and MST distance, as well as fiber orientation and dynamics. We then examined the human fetal cortex and found that a subset of human oRGs displayed similar characteristics, suggesting that diversity in oRG behavior may be a general feature. Similar to the human, ferret oRGs underwent multiple rounds of self-renewing divisions but were more likely to undergo symmetric divisions that expanded the oRG population, as opposed to producing intermediate progenitor cells (IPCs). Differences in oRG behaviors, including proliferative potential and daughter cell fates, may contribute to variations in cortical structure between mammalian species.
Collapse
|
37
|
Effects of allantoin on cognitive function and hippocampal neurogenesis. Food Chem Toxicol 2014; 64:210-6. [DOI: 10.1016/j.fct.2013.11.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 11/21/2013] [Accepted: 11/23/2013] [Indexed: 11/23/2022]
|
38
|
Abstract
To understand the emergence of human higher cognition, we must understand its biological substrate--the cerebral cortex, which considers itself the crowning achievement of evolution. Here, we describe how advances in developmental neurobiology, coupled with those in genetics, including adaptive protein evolution via gene duplications and the emergence of novel regulatory elements, can provide insights into the evolutionary mechanisms culminating in the human cerebrum. Given that the massive expansion of the cortical surface and elaboration of its connections in humans originates from developmental events, understanding the genetic regulation of cell number, neuronal migration to proper layers, columns, and regions, and ultimately their differentiation into specific phenotypes, is critical. The pre- and postnatal environment also interacts with the cellular substrate to yield a basic network that is refined via selection and elimination of synaptic connections, a process that is prolonged in humans. This knowledge provides essential insight into the pathogenesis of human-specific neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | | |
Collapse
|
39
|
Greig LC, Woodworth MB, Galazo MJ, Padmanabhan H, Macklis JD. Molecular logic of neocortical projection neuron specification, development and diversity. Nat Rev Neurosci 2013; 14:755-69. [PMID: 24105342 DOI: 10.1038/nrn3586] [Citation(s) in RCA: 583] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The sophisticated circuitry of the neocortex is assembled from a diverse repertoire of neuronal subtypes generated during development under precise molecular regulation. In recent years, several key controls over the specification and differentiation of neocortical projection neurons have been identified. This work provides substantial insight into the 'molecular logic' underlying cortical development and increasingly supports a model in which individual progenitor-stage and postmitotic regulators are embedded within highly interconnected networks that gate sequential developmental decisions. Here, we provide an integrative account of the molecular controls that direct the progressive development and delineation of subtype and area identity of neocortical projection neurons.
Collapse
|
40
|
Krubitzer L, Dooley JC. Cortical plasticity within and across lifetimes: how can development inform us about phenotypic transformations? Front Hum Neurosci 2013; 7:620. [PMID: 24130524 PMCID: PMC3793242 DOI: 10.3389/fnhum.2013.00620] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/08/2013] [Indexed: 11/13/2022] Open
Abstract
The neocortex is the part of the mammalian brain that is involved in perception, cognition, and volitional motor control. It is a highly dynamic structure that is dramatically altered within the lifetime of an animal and in different lineages throughout the course of evolution. These alterations account for the remarkable variations in behavior that species exhibit. Of particular interest is how these cortical phenotypes change within the lifetime of the individual and eventually evolve in species over time. Because we cannot study the evolution of the neocortex directly we use comparative analysis to appreciate the types of changes that have been made to the neocortex and the similarities that exist across taxa. Developmental studies inform us about how these phenotypic transitions may arise by alterations in developmental cascades or changes in the physical environment in which the brain develops. Both genes and the sensory environment contribute to aspects of the phenotype and similar features, such as the size of a cortical field, can be altered in a variety of ways. Although both genes and the laws of physics place constraints on the evolution of the neocortex, mammals have evolved a number of mechanisms that allow them to loosen these constraints and often alter the course of their own evolution.
Collapse
Affiliation(s)
- Leah Krubitzer
- Center for Neuroscience, University of California Davis, Davis, CA, USA ; Department of Psychology, University of California Davis, Davis, CA, USA
| | | |
Collapse
|
41
|
Wenger AM, Clarke SL, Notwell JH, Chung T, Tuteja G, Guturu H, Schaar BT, Bejerano G. The enhancer landscape during early neocortical development reveals patterns of dense regulation and co-option. PLoS Genet 2013; 9:e1003728. [PMID: 24009522 PMCID: PMC3757057 DOI: 10.1371/journal.pgen.1003728] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 07/03/2013] [Indexed: 11/18/2022] Open
Abstract
Genetic studies have identified a core set of transcription factors and target genes that control the development of the neocortex, the region of the human brain responsible for higher cognition. The specific regulatory interactions between these factors, many key upstream and downstream genes, and the enhancers that mediate all these interactions remain mostly uncharacterized. We perform p300 ChIP-seq to identify over 6,600 candidate enhancers active in the dorsal cerebral wall of embryonic day 14.5 (E14.5) mice. Over 95% of the peaks we measure are conserved to human. Eight of ten (80%) candidates tested using mouse transgenesis drive activity in restricted laminar patterns within the neocortex. GREAT based computational analysis reveals highly significant correlation with genes expressed at E14.5 in key areas for neocortex development, and allows the grouping of enhancers by known biological functions and pathways for further studies. We find that multiple genes are flanked by dozens of candidate enhancers each, including well-known key neocortical genes as well as suspected and novel genes. Nearly a quarter of our candidate enhancers are conserved well beyond mammals. Human and zebrafish regions orthologous to our candidate enhancers are shown to most often function in other aspects of central nervous system development. Finally, we find strong evidence that specific interspersed repeat families have contributed potentially key developmental enhancers via co-option. Our analysis expands the methodologies available for extracting the richness of information found in genome-wide functional maps.
Collapse
Affiliation(s)
- Aaron M. Wenger
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| | - Shoa L. Clarke
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - James H. Notwell
- Department of Computer Science, Stanford University, Stanford, California, United States of America
| | - Tisha Chung
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
| | - Geetu Tuteja
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
| | - Harendra Guturu
- Department of Electrical Engineering, Stanford University, Stanford, California, United States of America
| | - Bruce T. Schaar
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
| | - Gill Bejerano
- Department of Computer Science, Stanford University, Stanford, California, United States of America
- Department of Developmental Biology, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
Poluch S, Juliano SL. Fine-tuning of neurogenesis is essential for the evolutionary expansion of the cerebral cortex. ACTA ACUST UNITED AC 2013; 25:346-64. [PMID: 23968831 DOI: 10.1093/cercor/bht232] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We used several animal models to study global and regional cortical surface expansion: The lissencephalic mouse, gyrencephalic normal ferrets, in which the parietal cortex expands more than the temporal cortex, and moderately lissencephalic ferrets, showing a similar degree of temporal and parietal expansion. We found that overall cortical surface expansion is achieved when specific events occur prior to surpragranular layer formation. (1) The subventricular zone (SVZ) shows substantial growth, (2) the inner SVZ contains an increased number of outer radial glia and intermediate progenitor cells expressing Pax6, and (3) the outer SVZ contains a progenitor cell composition similar to the combined VZ and inner SVZ. A greater parietal expansion is also achieved by eliminating the latero-dorsal neurogenic gradient, so that neurogenesis displays a similar developmental degree between parietal and temporal regions. In contrast, mice or lissencephalic ferrets show more advanced neurogenesis in the temporal region. In conclusion, we propose that global and regional cortical surface expansion rely on similar strategies consisting in altering the timing of neurogenic events prior to the surpragranular layer formation, so that more progenitor cells, and ultimately more neurons, are produced. This hypothesis is supported by findings from a ferret model of lissencephaly obtained by transiently blocking neurogenesis during the formation of layer IV.
Collapse
Affiliation(s)
- Sylvie Poluch
- Department of Anatomy, Physiology, and Genetics Department of Neuroscience, Uniformed Services University, Bethesda, MD, USA
| | - Sharon L Juliano
- Department of Anatomy, Physiology, and Genetics Department of Neuroscience, Uniformed Services University, Bethesda, MD, USA
| |
Collapse
|
43
|
Tuoc TC, Narayanan R, Stoykova A. BAF chromatin remodeling complex: cortical size regulation and beyond. Cell Cycle 2013; 12:2953-9. [PMID: 23974113 DOI: 10.4161/cc.25999] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The multi-subunit chromatin remodeling BAF complex controls different developmental processes. Using cortex-specific conditional knockout and overexpression mouse models, we have recently reported that BAF170, a subunit of the vertebrate BAF chromatin remodeling complex, interacts with transcription factor (TF) Pax6 to control cortical size and volume. The mechanistic basis includes suppression of the expression of Pax6 target genes, which are required for genesis of cortical intermediate progenitors (IPs) and specification of late neuronal subtype identity. In addition, we showed that a dynamic competition between BAF170 and BAF155 subunits within the BAF complex during progression of neurogenesis is a primary event in modulating the size of the mammalian cortex. Here, we present additional insights into the interaction between the BAF complex and TF Pax6 in the genesis of IPs of the developing cortex. Furthermore, we show that such competition between BAF170 and BAF155 is involved as well in the determination of the size of the embryonic body. Our results add new insights into a cell-intrinsic mechanism, mediated by the chromatin remodeling BAF complex that controls vertebrate body shape and size.
Collapse
Affiliation(s)
- Tran Cong Tuoc
- Institute of Neuroanatomy; Universitätsmedizin Göttingen; Göttingen, Germany
| | | | | |
Collapse
|
44
|
Montiel JF, Kaune H, Maliqueo M. Maternal-fetal unit interactions and eutherian neocortical development and evolution. Front Neuroanat 2013; 7:22. [PMID: 23882189 PMCID: PMC3715729 DOI: 10.3389/fnana.2013.00022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 06/25/2013] [Indexed: 11/25/2022] Open
Abstract
The conserved brain design that primates inherited from early mammals differs from the variable adult brain size and species-specific brain dominances observed across mammals. This variability relies on the emergence of specialized cerebral cortical regions and sub-compartments, triggering an increase in brain size, areal interconnectivity and histological complexity that ultimately lies on the activation of developmental programs. Structural placental features are not well correlated with brain enlargement; however, several endocrine pathways could be tuned with the activation of neuronal progenitors in the proliferative neocortical compartments. In this article, we reviewed some mechanisms of eutherians maternal-fetal unit interactions associated with brain development and evolution. We propose a hypothesis of brain evolution where proliferative compartments in primates become activated by "non-classical" endocrine placental signals participating in different steps of corticogenesis. Changes in the inner placental structure, along with placenta endocrine stimuli over the cortical proliferative activity would allow mammalian brain enlargement with a concomitant shorter gestation span, as an evolutionary strategy to escape from parent-offspring conflict.
Collapse
Affiliation(s)
- Juan F. Montiel
- Centre for Biomedical Research, Facultad de Medicina, Universidad Diego PortalesSantiago, Chile.
| | - Heidy Kaune
- Centre for Biomedical Research, Facultad de Medicina, Universidad Diego PortalesSantiago, Chile.
- Nuffield Department of Obstetrics and Gynaecology, University of OxfordOxford, UK.
| | - Manuel Maliqueo
- Laboratorio de Endocrinología y Metabolismo, Departamento de Medicina Occidente, Facultad de Medicina, Universidad de ChileSantiago, Chile.
| |
Collapse
|
45
|
Kaas JH. The evolution of brains from early mammals to humans. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2012; 4:33-45. [PMID: 23529256 DOI: 10.1002/wcs.1206] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The large size and complex organization of the human brain makes it unique among primate brains. In particular, the neocortex constitutes about 80% of the brain, and this cortex is subdivided into a large number of functionally specialized regions, the cortical areas. Such a brain mediates accomplishments and abilities unmatched by any other species. How did such a brain evolve? Answers come from comparative studies of the brains of present-day mammals and other vertebrates in conjunction with information about brain sizes and shapes from the fossil record, studies of brain development, and principles derived from studies of scaling and optimal design. Early mammals were small, with small brains, an emphasis on olfaction, and little neocortex. Neocortex was transformed from the single layer of output pyramidal neurons of the dorsal cortex of earlier ancestors to the six layers of all present-day mammals. This small cap of neocortex was divided into 20-25 cortical areas, including primary and some of the secondary sensory areas that characterize neocortex in nearly all mammals today. Early placental mammals had a corpus callosum connecting the neocortex of the two hemispheres, a primary motor area, M1, and perhaps one or more premotor areas. One line of evolution, Euarchontoglires, led to present-day primates, tree shrews, flying lemurs, rodents, and rabbits. Early primates evolved from small-brained, nocturnal, insect-eating mammals with an expanded region of temporal visual cortex. These early nocturnal primates were adapted to the fine branch niche of the tropical rainforest by having an even more expanded visual system that mediated visually guided reaching and grasping of insects, small vertebrates, and fruits. Neocortex was greatly expanded and included an array of cortical areas that characterize neocortex of all living primates. Specializations of the visual system included new visual areas that contributed to a dorsal stream of visuomotor processing in a greatly enlarged region of posterior parietal cortex and an expanded motor system and the addition of a ventral premotor area. Higher visual areas in a large temporal lobe facilitated object recognition, and frontal cortex included granular prefrontal cortex. Auditory cortex included the primary and secondary auditory areas that characterize prosimian and anthropoid primates today. As anthropoids emerged as diurnal primates, the visual system specialized for detailed foveal vision. Other adaptations included an expansion of prefrontal cortex and insular cortex. The human and chimpanzee-bonobo lineages diverged some 6-8 million years ago with brains that were about one third the size of modern humans. Over the last 2 million years, the brains of our more recent ancestors increased greatly in size, especially in the prefrontal, posterior parietal, lateral temporal, and insular regions. Specialization of the two cerebral hemispheres for related, but different functions became pronounced, and language and other impressive cognitive abilities emerged. WIREs Cogn Sci 2013, 4:33-45. doi: 10.1002/wcs.1206 This article is categorized under: Neuroscience > Anatomy.
Collapse
Affiliation(s)
- Jon H Kaas
- Department of Psychology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
46
|
Dugas-Ford J, Rowell JJ, Ragsdale CW. Cell-type homologies and the origins of the neocortex. Proc Natl Acad Sci U S A 2012; 109:16974-9. [PMID: 23027930 PMCID: PMC3479531 DOI: 10.1073/pnas.1204773109] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The six-layered neocortex is a uniquely mammalian structure with evolutionary origins that remain in dispute. One long-standing hypothesis, based on similarities in neuronal connectivity, proposes that homologs of the layer 4 input and layer 5 output neurons of neocortex are present in the avian forebrain, where they contribute to specific nuclei rather than to layers. We devised a molecular test of this hypothesis based on layer-specific gene expression that is shared across rodent and carnivore neocortex. Our findings establish that the layer 4 input and the layer 5 output cell types are conserved across the amniotes, but are organized into very different architectures, forming nuclei in birds, cortical areas in reptiles, and cortical layers in mammals.
Collapse
Affiliation(s)
- Jennifer Dugas-Ford
- Department of Neurobiology and Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637
| | - Joanna J. Rowell
- Department of Neurobiology and Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637
| | - Clifton W. Ragsdale
- Department of Neurobiology and Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637
| |
Collapse
|
47
|
Tanaka DH, Nakajima K. GABAergic interneuron migration and the evolution of the neocortex. Dev Growth Differ 2012; 54:366-72. [PMID: 22524606 DOI: 10.1111/j.1440-169x.2012.01351.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A neocortex is present in all mammals but is not present in other classes of vertebrates, and the neocortex is extremely elaborate in humans. Changes in excitatory projection neurons and their progenitors within the developing dorsal pallium in the most recent common ancestor of mammals are thought to have been involved in the evolution of the neocortex. Our recent findings suggest that changes in the migratory ability of inhibitory interneurons derived from outside the neocortex may also have been involved in the evolution of the neocortex. In this article we review the literature on the migratory profile of inhibitory interneurons in several different species and the literature on comparisons between the intrinsic migratory ability of interneurons derived from different species. Finally, we propose a hypothesis about the mammalian-specific evolution of the migratory ability of interneurons and its potential contribution to the establishment of a functional neocortex.
Collapse
Affiliation(s)
- Daisuke H Tanaka
- Department of Anatomy, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | | |
Collapse
|
48
|
Pacini S, Fiore MG, Magherini S, Morucci G, Branca JJV, Gulisano M, Ruggiero M. Could cadmium be responsible for some of the neurological signs and symptoms of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Med Hypotheses 2012; 79:403-7. [PMID: 22795611 DOI: 10.1016/j.mehy.2012.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 06/04/2012] [Accepted: 06/17/2012] [Indexed: 01/29/2023]
Abstract
According to the World Health Organization, Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a neurological disease characterized by widespread inflammation and multi-systemic neuropathology. Aetiology and pathogenesis are unknown, and several agents have been proposed as causative agents or as factors perpetuating the syndrome. Exposure to heavy metals, with particular reference to mercury and gold in dental amalgams, has been considered among the triggers of ME/CFS. Here we hypothesize that cadmium, a widespread occupational and environmental heavy metal pollutant, might be associated with some of the neurological findings described in ME/CFS. In fact, ME/CFS patients show a decrease of the volume of the gray matter in turn associated with objective reduction of physical activity. Cadmium induces neuronal death in cortical neurons through a combined mechanism of apoptosis and necrosis and it could then be hypothesized that cadmium-induced neuronal cell death is responsible for some of the effects of cadmium on the central nervous system, i.e. a decrease in attention level and memory in exposed humans as well as to a diminished ability for training and learning in rats, that are symptoms typical of ME/CFS. This hypothesis can be tested by measuring cadmium exposure in a cohort of ME/CFS patients compared with matched healthy controls, and by measuring gray matter volume in un-exposed healthy controls, exposed non-ME/CFS subjects, un-exposed ME/CFS patients and exposed ME/CFS patients. In addition, we hypothesize that cadmium exposure could be associated with reduced cerebral blood flow in ME/CFS patients because of the disruptive effects of cadmium on angiogenesis. In fact, cadmium inhibits angiogenesis and low global cerebral flow is associated with abnormal brain neuroimaging results and brain dysfunction in the form of reduced cognitive testing scores in ME/CFS patients. This hypothesis can be tested by measuring cerebral cortex blood flow in un-exposed healthy controls, exposed non-ME/CFS subjects, un-exposed ME/CFS patients and exposed ME/CFS patients. If our hypothesis is demonstrated correct, the consequences could affect prevention, early diagnosis, and treatment of ME/CFS. Implications in early diagnosis could entail the evaluation of symptoms typical of ME/CFS in cadmium-exposed subjects as well as the search for signs of exposure to cadmium in subjects diagnosed with ME/CFS. Nutritional supplementation of magnesium and zinc could then be considered, since these elements have been proposed in the prophylaxis and therapy of cadmium exposure, and magnesium was demonstrated effective on ME/CFS patients' symptom profiles.
Collapse
Affiliation(s)
- Stefania Pacini
- Department of Anatomy, Histology and Forensic Medicine, University of Firenze, Viale Morgagni 85, 50134 Firenze, Italy.
| | | | | | | | | | | | | |
Collapse
|
49
|
Transcriptomes of germinal zones of human and mouse fetal neocortex suggest a role of extracellular matrix in progenitor self-renewal. Proc Natl Acad Sci U S A 2012; 109:11836-41. [PMID: 22753484 DOI: 10.1073/pnas.1209647109] [Citation(s) in RCA: 237] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The expansion of the neocortex during mammalian brain evolution results primarily from an increase in neural progenitor cell divisions in its two principal germinal zones during development, the ventricular zone (VZ) and the subventricular zone (SVZ). Using mRNA sequencing, we analyzed the transcriptomes of fetal human and embryonic mouse VZ, SVZ, and cortical plate. In mouse, the transcriptome of the SVZ was more similar to that of the cortical plate than that of the VZ, whereas in human the opposite was the case, with the inner and outer SVZ being highly related to each other despite their cytoarchitectonic differences. We describe sets of genes that are up- or down-regulated in each germinal zone. These data suggest that cell adhesion and cell-extracellular matrix interactions promote the proliferation and self-renewal of neural progenitors in the developing human neocortex. Notably, relevant extracellular matrix-associated genes include distinct sets of collagens, laminins, proteoglycans, and integrins, along with specific sets of growth factors and morphogens. Our data establish a basis for identifying novel cell-type markers and open up avenues to unravel the molecular basis of neocortex expansion during evolution.
Collapse
|
50
|
|