1
|
LaNoce E, Zhang DY, Garcia-Epelboim A, Su Y, Sun Y, Alepa G, Angelucci AR, Akay-Espinoza C, Jordan-Sciutto KL, Song H, Ming GL, Christian KM. Exposure to the antiretroviral drug dolutegravir impairs structure and neurogenesis in a forebrain organoid model of human embryonic cortical development. Front Mol Neurosci 2024; 17:1459877. [PMID: 39569018 PMCID: PMC11576471 DOI: 10.3389/fnmol.2024.1459877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/13/2024] [Indexed: 11/22/2024] Open
Abstract
Introduction For many therapeutic drugs, including antiretroviral drugs used to treat people living with HIV-1 (PLWH), we have little data on the potential effects on the developing human brain due to limited access to tissue and historical constraints on the inclusion of pregnant populations in clinical trials. Human induced pluripotent stem cells (iPSCs) offer a new avenue to gain insight on how drugs may impact human cell types representative of the developing central nervous system. To prevent vertical transmission of HIV and promote the health of pregnant PLWH, antiretroviral therapy must be initiated and/or maintained throughout pregnancy. However, many antiretroviral drugs are approved for widespread use following clinical testing only in non-pregnant populations and there may be limited information on potential teratogenicity until pregnancy outcomes are evaluated. The integrase strand transfer inhibitor dolutegravir (DTG) is a frontline antiretroviral drug that is effective in viral suppression of HIV but was previously reported to be associated with a slight increase in the risk for neural tube defects in one study, although this has not been replicated in other cohorts. Methods To directly investigate the potential impact of DTG on human cortical neurogenesis, we measured the effects of daily drug exposure on the early stages of corticogenesis in a human iPSC-based forebrain organoid model. We quantified organoid size and structure and analyzed gene and protein expression to evaluate the impact of several doses of DTG on organoid development. Results We observed deficits in organoid structure and impaired neurogenesis in DTG-treated organoids compared to vehicle-treated control organoids after 20 or 40 days in culture. Our highest dose of DTG (10 μM) resulted in significantly smaller organoids with a reduced density of neural rosette structures compared to vehicle-treated controls. Mechanistically, RNA-sequencing and immunohistological analysis suggests dysregulated amino acid transport and activation of the integrated stress response in the DTG-treated organoids, and functionally, a small molecule integrated stress response inhibitor (ISRIB) could partially rescue increased expression of proteins related to cell cycle regulation. Discussion Together, these results illustrate the potential for human iPSC-based strategies to reveal biological processes during neurogenesis that may be affected by therapeutic drugs and provide complementary data in relevant human cell types to augment preclinical investigations of drug safety during pregnancy.
Collapse
Affiliation(s)
- Emma LaNoce
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Daniel Y. Zhang
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alan Garcia-Epelboim
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yusha Sun
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Giana Alepa
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Angelina R. Angelucci
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cagla Akay-Espinoza
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kelly L. Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kimberly M. Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
2
|
Luu P, Tucker DM. Continuity and change in neural plasticity through embryonic morphogenesis, fetal activity-dependent synaptogenesis, and infant memory consolidation. Dev Psychobiol 2023; 65:e22439. [PMID: 38010309 DOI: 10.1002/dev.22439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/09/2023] [Accepted: 10/18/2023] [Indexed: 11/29/2023]
Abstract
There is an apparent continuity in human neural development that can be traced to venerable themes of vertebrate morphogenesis that have shaped the evolution of the reptilian telencephalon (including both primitive three-layered cortex and basal ganglia) and then the subsequent evolution of the mammalian six-layered neocortex. In this theoretical analysis, we propose that an evolutionary-developmental analysis of these general morphogenetic themes can help to explain the embryonic development of the dual divisions of the limbic system that control the dorsal and ventral networks of the human neocortex. These include the archicortical (dorsal limbic) Papez circuits regulated by the hippocampus that organize spatial, contextual memory, as well as the paleocortical (ventral limbic) circuits that organize object memory. We review evidence that these dorsal and ventral limbic divisions are controlled by the differential actions of brainstem lemnothalamic and midbrain collothalamic arousal control systems, respectively, thereby traversing the vertebrate subcortical neuraxis. These dual control systems are first seen shaping the phyletic morphogenesis of the archicortical and paleocortical foundations of the forebrain in embryogenesis. They then provide dual modes of activity-dependent synaptic organization in the active (lemnothalamic) and quiet (collothalamic) stages of fetal sleep. Finally, these regulatory systems mature to form the major systems of memory consolidation of postnatal development, including the rapid eye movement (lemnothalamic) consolidation of implicit memory and social attachment in the first year, and then-in a subsequent stage-the non-REM (collothalamic) consolidation of explicit memory that is integral to the autonomy and individuation of the second year of life.
Collapse
Affiliation(s)
- Phan Luu
- Brain Electrophysiology Laboratory Company, Eugene, Oregon, USA
- Department of Psychology, University of Oregon, Eugene, Oregon, USA
| | - Don M Tucker
- Brain Electrophysiology Laboratory Company, Eugene, Oregon, USA
- Department of Psychology, University of Oregon, Eugene, Oregon, USA
| |
Collapse
|
3
|
Berdugo‐Vega G, Dhingra S, Calegari F. Sharpening the blades of the dentate gyrus: how adult-born neurons differentially modulate diverse aspects of hippocampal learning and memory. EMBO J 2023; 42:e113524. [PMID: 37743770 PMCID: PMC11059975 DOI: 10.15252/embj.2023113524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
For decades, the mammalian hippocampus has been the focus of cellular, anatomical, behavioral, and computational studies aimed at understanding the fundamental mechanisms underlying cognition. Long recognized as the brain's seat for learning and memory, a wealth of knowledge has been accumulated on how the hippocampus processes sensory input, builds complex associations between objects, events, and space, and stores this information in the form of memories to be retrieved later in life. However, despite major efforts, our understanding of hippocampal cognitive function remains fragmentary, and models trying to explain it are continually revisited. Here, we review the literature across all above-mentioned domains and offer a new perspective by bringing attention to the most distinctive, and generally neglected, feature of the mammalian hippocampal formation, namely, the structural separability of the two blades of the dentate gyrus into "supra-pyramidal" and "infra-pyramidal". Next, we discuss recent reports supporting differential effects of adult neurogenesis in the regulation of mature granule cell activity in these two blades. We propose a model for how differences in connectivity and adult neurogenesis in the two blades can potentially provide a substrate for subtly different cognitive functions.
Collapse
Affiliation(s)
- Gabriel Berdugo‐Vega
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
- Present address:
Laboratory of Neuroepigenetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale Lausanne (EPFL)LausanneSwitzerland
| | - Shonali Dhingra
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
| | - Federico Calegari
- CRTD‐Center for Regenerative Therapies DresdenTechnische Universität DresdenDresdenGermany
| |
Collapse
|
4
|
Krienen FM, Levandowski KM, Zaniewski H, del Rosario RC, Schroeder ME, Goldman M, Wienisch M, Lutservitz A, Beja-Glasser VF, Chen C, Zhang Q, Chan KY, Li KX, Sharma J, McCormack D, Shin TW, Harrahill A, Nyase E, Mudhar G, Mauermann A, Wysoker A, Nemesh J, Kashin S, Vergara J, Chelini G, Dimidschstein J, Berretta S, Deverman BE, Boyden E, McCarroll SA, Feng G. A marmoset brain cell census reveals regional specialization of cellular identities. SCIENCE ADVANCES 2023; 9:eadk3986. [PMID: 37824615 PMCID: PMC10569717 DOI: 10.1126/sciadv.adk3986] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023]
Abstract
The mammalian brain is composed of many brain structures, each with its own ontogenetic and developmental history. We used single-nucleus RNA sequencing to sample over 2.4 million brain cells across 18 locations in the common marmoset, a New World monkey primed for genetic engineering, and examined gene expression patterns of cell types within and across brain structures. The adult transcriptomic identity of most neuronal types is shaped more by developmental origin than by neurotransmitter signaling repertoire. Quantitative mapping of GABAergic types with single-molecule FISH (smFISH) reveals that interneurons in the striatum and neocortex follow distinct spatial principles, and that lateral prefrontal and other higher-order cortical association areas are distinguished by high proportions of VIP+ neurons. We use cell type-specific enhancers to drive AAV-GFP and reconstruct the morphologies of molecularly resolved interneuron types in neocortex and striatum. Our analyses highlight how lineage, local context, and functional class contribute to the transcriptional identity and biodistribution of primate brain cell types.
Collapse
Affiliation(s)
- Fenna M. Krienen
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kirsten M. Levandowski
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Heather Zaniewski
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ricardo C.H. del Rosario
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Margaret E. Schroeder
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Melissa Goldman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Martin Wienisch
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alyssa Lutservitz
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Victoria F. Beja-Glasser
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Cindy Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Qiangge Zhang
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ken Y. Chan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Katelyn X. Li
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jitendra Sharma
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Dana McCormack
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Tay Won Shin
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| | - Andrew Harrahill
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eric Nyase
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gagandeep Mudhar
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA
| | - Abigail Mauermann
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| | - Alec Wysoker
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - James Nemesh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Seva Kashin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Josselyn Vergara
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gabriele Chelini
- Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura n.1, Rovereto (TN) 38068, Italy
| | - Jordane Dimidschstein
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sabina Berretta
- Basic Neuroscience Division, McLean Hospital, Belmont, MA 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Benjamin E. Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ed Boyden
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Cambridge, MA 02139, USA
| | - Steven A. McCarroll
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Guoping Feng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
5
|
Ma Y, Zhou X. Spatially informed cell-type deconvolution for spatial transcriptomics. Nat Biotechnol 2022; 40:1349-1359. [PMID: 35501392 PMCID: PMC9464662 DOI: 10.1038/s41587-022-01273-7] [Citation(s) in RCA: 210] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 03/07/2022] [Indexed: 12/16/2022]
Abstract
Many spatially resolved transcriptomic technologies do not have single-cell resolution but measure the average gene expression for each spot from a mixture of cells of potentially heterogeneous cell types. Here, we introduce a deconvolution method, conditional autoregressive-based deconvolution (CARD), that combines cell-type-specific expression information from single-cell RNA sequencing (scRNA-seq) with correlation in cell-type composition across tissue locations. Modeling spatial correlation allows us to borrow the cell-type composition information across locations, improving accuracy of deconvolution even with a mismatched scRNA-seq reference. CARD can also impute cell-type compositions and gene expression levels at unmeasured tissue locations to enable the construction of a refined spatial tissue map with a resolution arbitrarily higher than that measured in the original study and can perform deconvolution without an scRNA-seq reference. Applications to four datasets, including a pancreatic cancer dataset, identified multiple cell types and molecular markers with distinct spatial localization that define the progression, heterogeneity and compartmentalization of pancreatic cancer.
Collapse
Affiliation(s)
- Ying Ma
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Nano PR, Bhaduri A. Evaluation of advances in cortical development using model systems. Dev Neurobiol 2022; 82:408-427. [PMID: 35644985 PMCID: PMC10924780 DOI: 10.1002/dneu.22879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 11/11/2022]
Abstract
Compared with that of even the closest primates, the human cortex displays a high degree of specialization and expansion that largely emerges developmentally. Although decades of research in the mouse and other model systems has revealed core tenets of cortical development that are well preserved across mammalian species, small deviations in transcription factor expression, novel cell types in primates and/or humans, and unique cortical architecture distinguish the human cortex. Importantly, many of the genes and signaling pathways thought to drive human-specific cortical expansion also leave the brain vulnerable to disease, as the misregulation of these factors is highly correlated with neurodevelopmental and neuropsychiatric disorders. However, creating a comprehensive understanding of human-specific cognition and disease remains challenging. Here, we review key stages of cortical development and highlight known or possible differences between model systems and the developing human brain. By identifying the developmental trajectories that may facilitate uniquely human traits, we highlight open questions in need of approaches to examine these processes in a human context and reveal translatable insights into human developmental disorders.
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
7
|
López-Mengual A, Segura-Feliu M, Sunyer R, Sanz-Fraile H, Otero J, Mesquida-Veny F, Gil V, Hervera A, Ferrer I, Soriano J, Trepat X, Farré R, Navajas D, Del Río JA. Involvement of Mechanical Cues in the Migration of Cajal-Retzius Cells in the Marginal Zone During Neocortical Development. Front Cell Dev Biol 2022; 10:886110. [PMID: 35652101 PMCID: PMC9150848 DOI: 10.3389/fcell.2022.886110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence points to coordinated action of chemical and mechanical cues during brain development. At early stages of neocortical development, angiogenic factors and chemokines such as CXCL12, ephrins, and semaphorins assume crucial roles in orchestrating neuronal migration and axon elongation of postmitotic neurons. Here we explore the intrinsic mechanical properties of the developing marginal zone of the pallium in the migratory pathways and brain distribution of the pioneer Cajal-Retzius cells. These neurons are generated in several proliferative regions in the developing brain (e.g., the cortical hem and the pallial subpallial boundary) and migrate tangentially in the preplate/marginal zone covering the upper portion of the developing cortex. These cells play crucial roles in correct neocortical layer formation by secreting several molecules such as Reelin. Our results indicate that the motogenic properties of Cajal-Retzius cells and their perinatal distribution in the marginal zone are modulated by both chemical and mechanical factors, by the specific mechanical properties of Cajal-Retzius cells, and by the differential stiffness of the migratory routes. Indeed, cells originating in the cortical hem display higher migratory capacities than those generated in the pallial subpallial boundary which may be involved in the differential distribution of these cells in the dorsal-lateral axis in the developing marginal zone.
Collapse
Affiliation(s)
- Ana López-Mengual
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Miriam Segura-Feliu
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Raimon Sunyer
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Héctor Sanz-Fraile
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Jorge Otero
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain
| | - Francina Mesquida-Veny
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Arnau Hervera
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Isidre Ferrer
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Senior Consultant, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Jordi Soriano
- Departament de Física de La Matèria Condensada, Universitat de Barcelona, Barcelona, Spain.,University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Xavier Trepat
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Integrative Cell and Tissue Dynamics, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain.,Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Institució Catalana de Recerca I Estudis Avançats, University of Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain.,Institut D'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain.,Cellular and Respiratory Biomechanics, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| |
Collapse
|
8
|
Jiménez S, Moreno N. Analysis of the Expression Pattern of Cajal-Retzius Cell Markers in the Xenopus laevis Forebrain. BRAIN, BEHAVIOR AND EVOLUTION 2021; 96:263-282. [PMID: 34614492 DOI: 10.1159/000519025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/09/2021] [Indexed: 01/26/2023]
Abstract
Cajal-Retzius cells are essential for cortical development in mammals, and their involvement in the evolution of this structure has been widely postulated, but very little is known about their progenitor domains in non-mammalian vertebrates. Using in situhybridization and immunofluorescence techniques we analyzed the expression of some of the main Cajal-Retzius cell markers such as Dbx1, Ebf3, ER81, Lhx1, Lhx5, p73, Reelin, Wnt3a, Zic1, and Zic2 in the forebrain of the anuran Xenopus laevis, because amphibians are the only class of anamniote tetrapods and show a tetrapartite evaginated pallium, but no layered or nuclear organization. Our results suggested that the Cajal-Retzius cell progenitor domains were comparable to those previously described in amniotes. Thus, at dorsomedial telencephalic portions a region comparable to the cortical hem was defined in Xenopus based on the expression of Wnt3a, p73, Reelin, Zic1, and Zic2. In the septum, two different domains were observed: a periventricular dorsal septum, at the limit between the pallium and the subpallium, expressing Reelin, Zic1, and Zic2, and a related septal domain, expressing Ebf3, Zic1, and Zic2. In the lateral telencephalon, the ventral pallium next to the pallio-subpallial boundary, the lack of Dbx1 and the unique expression of Reelin during development defined this territory as the most divergent with respect to mammals. Finally, we also analyzed the expression of these markers at the prethalamic eminence region, suggested as Cajal-Retzius progenitor domain in amniotes, observing there Zic1, Zic2, ER81, and Lhx1 expression. Our data show that in anurans there are different subtypes and progenitor domains of Cajal-Retzius cells, which probably contribute to the cortical regional specification and territory-specific properties. This supports the notion that the basic organization of pallial derivatives in vertebrates follows a comparable fundamental arrangement, even in those that do not have a sophisticated stratified cortical structure like the mammalian cerebral cortex.
Collapse
Affiliation(s)
- Sara Jiménez
- Department of Cell Biology, Faculty of Biology, University Complutense, Madrid, Spain
| | - Nerea Moreno
- Department of Cell Biology, Faculty of Biology, University Complutense, Madrid, Spain
| |
Collapse
|
9
|
Sarieva K, Mayer S. The Effects of Environmental Adversities on Human Neocortical Neurogenesis Modeled in Brain Organoids. Front Mol Biosci 2021; 8:686410. [PMID: 34250020 PMCID: PMC8264783 DOI: 10.3389/fmolb.2021.686410] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past decades, a growing body of evidence has demonstrated the impact of prenatal environmental adversity on the development of the human embryonic and fetal brain. Prenatal environmental adversity includes infectious agents, medication, and substances of use as well as inherently maternal factors, such as diabetes and stress. These adversities may cause long-lasting effects if occurring in sensitive time windows and, therefore, have high clinical relevance. However, our knowledge of their influence on specific cellular and molecular processes of in utero brain development remains scarce. This gap of knowledge can be partially explained by the restricted experimental access to the human embryonic and fetal brain and limited recapitulation of human-specific neurodevelopmental events in model organisms. In the past years, novel 3D human stem cell-based in vitro modeling systems, so-called brain organoids, have proven their applicability for modeling early events of human brain development in health and disease. Since their emergence, brain organoids have been successfully employed to study molecular mechanisms of Zika and Herpes simplex virus-associated microcephaly, as well as more subtle events happening upon maternal alcohol and nicotine consumption. These studies converge on pathological mechanisms targeting neural stem cells. In this review, we discuss how brain organoids have recently revealed commonalities and differences in the effects of environmental adversities on human neurogenesis. We highlight both the breakthroughs in understanding the molecular consequences of environmental exposures achieved using organoids as well as the on-going challenges in the field related to variability in protocols and a lack of benchmarking, which make cross-study comparisons difficult.
Collapse
Affiliation(s)
- Kseniia Sarieva
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- International Max Planck Research School, Graduate Training Centre of Neuroscience, University of Tübingen, Tübingen, Germany
| | - Simone Mayer
- Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| |
Collapse
|
10
|
Moore SA, Iulianella A. Development of the mammalian cortical hem and its derivatives: the choroid plexus, Cajal-Retzius cells and hippocampus. Open Biol 2021; 11:210042. [PMID: 33947245 PMCID: PMC8097212 DOI: 10.1098/rsob.210042] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/08/2021] [Indexed: 12/14/2022] Open
Abstract
The dorsal medial region of the developing mammalian telencephalon plays a central role in the patterning of the adjacent brain regions. This review describes the development of this specialized region of the vertebrate brain, called the cortical hem, and the formation of the various cells and structures it gives rise to, including the choroid plexus, Cajal-Retzius cells and the hippocampus. We highlight the ontogenic processes that create these different forebrain derivatives from their shared embryonic origin and discuss the key signalling pathways and molecules that influence the patterning of the cortical hem. These include BMP, Wnt, FGF and Shh signalling pathways acting with Homeobox factors to carve the medial telencephalon into district progenitor regions, which in turn give rise to the choroid plexus, dentate gyrus and hippocampus. We then link the formation of the lateral ventricle choroid plexus with embryonic and postnatal neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Samantha A. Moore
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia, Canada, B3H4R2
| | - Angelo Iulianella
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, and Brain Repair Centre, Life Science Research Institute, 1348 Summer Street, Halifax, Nova Scotia, Canada, B3H4R2
| |
Collapse
|
11
|
Li J, Sun L, Peng XL, Yu XM, Qi SJ, Lu ZJ, Han JDJ, Shen Q. Integrative genomic analysis of early neurogenesis reveals a temporal genetic program for differentiation and specification of preplate and Cajal-Retzius neurons. PLoS Genet 2021; 17:e1009355. [PMID: 33760820 PMCID: PMC7990179 DOI: 10.1371/journal.pgen.1009355] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 01/12/2021] [Indexed: 01/02/2023] Open
Abstract
Neurogenesis in the developing neocortex begins with the generation of the preplate, which consists of early-born neurons including Cajal-Retzius (CR) cells and subplate neurons. Here, utilizing the Ebf2-EGFP transgenic mouse in which EGFP initially labels the preplate neurons then persists in CR cells, we reveal the dynamic transcriptome profiles of early neurogenesis and CR cell differentiation. Genome-wide RNA-seq and ChIP-seq analyses at multiple early neurogenic stages have revealed the temporal gene expression dynamics of early neurogenesis and distinct histone modification patterns in early differentiating neurons. We have identified a new set of coding genes and lncRNAs involved in early neuronal differentiation and validated with functional assays in vitro and in vivo. In addition, at E15.5 when Ebf2-EGFP+ cells are mostly CR neurons, single-cell sequencing analysis of purified Ebf2-EGFP+ cells uncovers molecular heterogeneities in CR neurons, but without apparent clustering of cells with distinct regional origins. Along a pseudotemporal trajectory these cells are classified into three different developing states, revealing genetic cascades from early generic neuronal differentiation to late fate specification during the establishment of CR neuron identity and function. Our findings shed light on the molecular mechanisms governing the early differentiation steps during cortical development, especially CR neuron differentiation. Neural stem cells and progenitor cells in the embryonic brain give rise to neurons following a precise temporal order after initial expansion. Early-born neurons including Cajal-Retzius (CR) cells and subplate neurons form the preplate in the developing cerebral cortex, then CR neurons occupy the layer 1, playing an important role in cortical histogenesis. The molecular mechanisms governing the early neuronal differentiation processes remain to be explored. Here, by genome-wide approaches including bulk RNA-seq, single-cell RNA-seq and ChIP-seq, we comprehensively characterized the temporal dynamic gene expression profile and epigenetic status at different stages during early cortical development and uncovered molecularly heterogeneous subpopulations within the CR cells. We revealed CR neuron signatures and cell type-specific histone modification patterns along early neuron specification. Using in vitro and in vivo assays, we identified novel lncRNAs as potential functional regulators in preplate differentiation and CR neuron identity establishment. Our study provides a comprehensive analysis of the genetic and epigenetic programs during neuronal differentiation and would help bring new insights into the early cortical neurogenesis process, particularly the differentiation of CR neurons.
Collapse
Affiliation(s)
- Jia Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- PTN graduate program, School of Life Sciences, Peking University, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Lei Sun
- PTN graduate program, School of Life Sciences, Tsinghua University, Beijing, China
| | | | - Xiao-Ming Yu
- School of Medicine, Tsinghua University, Beijing, China
| | - Shao-Jun Qi
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Zhi John Lu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jing-Dong J. Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qin Shen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Ministry of Education, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Brain and Spinal Cord Clinical Research Center, Tongji University, Shanghai, China
- * E-mail:
| |
Collapse
|
12
|
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: 15] [Impact Index Per Article: 3.8] [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
|
13
|
Puelles L, Diaz C, Stühmer T, Ferran JL, Martínez‐de la Torre M, Rubenstein JLR. LacZ-reporter mapping of Dlx5/6 expression and genoarchitectural analysis of the postnatal mouse prethalamus. J Comp Neurol 2021; 529:367-420. [PMID: 32420617 PMCID: PMC7671952 DOI: 10.1002/cne.24952] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 05/10/2020] [Accepted: 05/11/2020] [Indexed: 12/22/2022]
Abstract
We present here a thorough and complete analysis of mouse P0-P140 prethalamic histogenetic subdivisions and corresponding nuclear derivatives, in the context of local tract landmarks. The study used as fundamental material brains from a transgenic mouse line that expresses LacZ under the control of an intragenic enhancer of Dlx5 and Dlx6 (Dlx5/6-LacZ). Subtle shadings of LacZ signal, jointly with pan-DLX immunoreaction, and several other ancillary protein or RNA markers, including Calb2 and Nkx2.2 ISH (for the prethalamic eminence, and derivatives of the rostral zona limitans shell domain, respectively) were mapped across the prethalamus. The resulting model of the prethalamic region postulates tetrapartite rostrocaudal and dorsoventral subdivisions, as well as a tripartite radial stratification, each cell population showing a characteristic molecular profile. Some novel nuclei are proposed, and some instances of potential tangential cell migration were noted.
Collapse
Affiliation(s)
- Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB‐Arrixaca InstituteUniversity of MurciaMurciaSpain
| | - Carmen Diaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological DisabilitiesUniversity of Castilla‐La ManchaAlbaceteSpain
| | - Thorsten Stühmer
- Nina Ireland Laboratory of Developmental Neurobiology, Department of PsychiatryUCSF Medical SchoolSan FranciscoCaliforniaUSA
| | - José L. Ferran
- Department of Human Anatomy and Psychobiology and IMIB‐Arrixaca InstituteUniversity of MurciaMurciaSpain
| | | | - John L. R. Rubenstein
- Nina Ireland Laboratory of Developmental Neurobiology, Department of PsychiatryUCSF Medical SchoolSan FranciscoCaliforniaUSA
| |
Collapse
|
14
|
Fan X, Fu Y, Zhou X, Sun L, Yang M, Wang M, Chen R, Wu Q, Yong J, Dong J, Wen L, Qiao J, Wang X, Tang F. Single-cell transcriptome analysis reveals cell lineage specification in temporal-spatial patterns in human cortical development. SCIENCE ADVANCES 2020; 6:eaaz2978. [PMID: 32923614 PMCID: PMC7450478 DOI: 10.1126/sciadv.aaz2978] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 07/09/2020] [Indexed: 05/19/2023]
Abstract
Neurogenesis processes differ in different areas of the cortex in many species, including humans. Here, we performed single-cell transcriptome profiling of the four cortical lobes and pons during human embryonic and fetal development. We identified distinct subtypes of neural progenitor cells (NPCs) and their molecular signatures, including a group of previously unidentified transient NPCs. We specified the neurogenesis path and molecular regulations of the human deep-layer, upper-layer, and mature neurons. Neurons showed clear spatial and temporal distinctions, while glial cells of different origins showed development patterns similar to those of mice, and we captured the developmental trajectory of oligodendrocyte lineage cells until the human mid-fetal stage. Additionally, we verified region-specific characteristics of neurons in the cortex, including their distinct electrophysiological features. With systematic single-cell analysis, we decoded human neuronal development in temporal and spatial dimensions from GW7 to GW28, offering deeper insights into the molecular regulations underlying human neurogenesis and cortical development.
Collapse
Affiliation(s)
- Xiaoying Fan
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Biomedical Pioneering Innovation Center and Center for Reproductive Medicine, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Brain-Intelligence Science and Technology, Zhangjing Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanyuan Fu
- Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xin Zhou
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Brain-Intelligence Science and Technology, Zhangjing Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Le Sun
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Brain-Intelligence Science and Technology, Zhangjing Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Yang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
| | - Mengdi Wang
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ruiguo Chen
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Brain-Intelligence Science and Technology, Zhangjing Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Wu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Brain-Intelligence Science and Technology, Zhangjing Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Yong
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
| | - Ji Dong
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Biomedical Pioneering Innovation Center and Center for Reproductive Medicine, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Lu Wen
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Biomedical Pioneering Innovation Center and Center for Reproductive Medicine, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
| | - Jie Qiao
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing 100191, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology (Shanghai), Institute of Brain-Intelligence Science and Technology, Zhangjing Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Advanced Innovation Center for Human Brain Protection, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Fuchou Tang
- Beijing Advanced Innovation Center for Genomics, Department of Obstetrics and Gynecology, College of Life Sciences, Third Hospital, Peking University, Beijing 100871, China
- Biomedical Pioneering Innovation Center and Center for Reproductive Medicine, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
15
|
Abstract
The p53 family member p73 has a complex gene structure, including alternative promoters and alternative splicing of the 3' UTR. This results in a complex range of isoforms whose biological relevance largely remains to be determined. By deleting exon 13 (which encodes a sterile α motif) from the Trp73 gene, we selectively engineered mice to replace the most abundantly expressed C-terminal isoform, p73α, with a shorter product of alternative splicing, p73β. These mice (Trp73 Δ13/Δ13 ) display severe neurodevelopmental defects with significant functional and morphological abnormalities. Replacement of p73α with p73β results in the depletion of Cajal-Retzius (CR) cells in embryonic stages, thus depriving the developing hippocampus of the pool of neurons necessary for correct hippocampal architecture. Consequently, Trp73 Δ13/Δ13 mice display severe hippocampal dysgenesis, reduced synaptic functionality and impaired learning and memory capabilities. Our data shed light on the relevance of p73 alternative splicing and show that the full-length C terminus of p73 is essential for hippocampal development.
Collapse
|
16
|
Longitudinal developmental analysis of prethalamic eminence derivatives in the chick by mapping of Tbr1 in situ expression. Brain Struct Funct 2020; 225:481-510. [DOI: 10.1007/s00429-019-02015-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023]
|
17
|
Kast RJ, Levitt P. Precision in the development of neocortical architecture: From progenitors to cortical networks. Prog Neurobiol 2019; 175:77-95. [PMID: 30677429 PMCID: PMC6402587 DOI: 10.1016/j.pneurobio.2019.01.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/02/2019] [Accepted: 01/21/2019] [Indexed: 02/07/2023]
Abstract
Of all brain regions, the 6-layered neocortex has undergone the most dramatic changes in size and complexity during mammalian brain evolution. These changes, occurring in the context of a conserved set of organizational features that emerge through stereotypical developmental processes, are considered responsible for the cognitive capacities and sensory specializations represented within the mammalian clade. The modern experimental era of developmental neurobiology, spanning 6 decades, has deciphered a number of mechanisms responsible for producing the diversity of cortical neuron types, their precise connectivity and the role of gene by environment interactions. Here, experiments providing insight into the development of cortical projection neuron differentiation and connectivity are reviewed. This current perspective integrates discussion of classic studies and new findings, based on recent technical advances, to highlight an improved understanding of the neuronal complexity and precise connectivity of cortical circuitry. These descriptive advances bring new opportunities for studies related to the developmental origins of cortical circuits that will, in turn, improve the prospects of identifying pathogenic targets of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ryan J Kast
- Department of Pediatrics and Program in Developmental Neuroscience and Developmental Neurogenetics, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, USA
| | - Pat Levitt
- Department of Pediatrics and Program in Developmental Neuroscience and Developmental Neurogenetics, The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90027, USA.
| |
Collapse
|
18
|
Holst CB, Brøchner CB, Vitting-Seerup K, Møllgård K. Astrogliogenesis in human fetal brain: complex spatiotemporal immunoreactivity patterns of GFAP, S100, AQP4 and YKL-40. J Anat 2019; 235:590-615. [PMID: 30901080 DOI: 10.1111/joa.12948] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
The astroglial lineage consists of heterogeneous cells instrumental for normal brain development, function and repair. Unfortunately, this heterogeneity complicates research in the field, which suffers from lack of truly specific and sensitive astroglial markers. Nevertheless, single astroglial markers are often used to describe astrocytes in different settings. We therefore investigated and compared spatiotemporal patterns of immunoreactivity in developing human brain from 12 to 21 weeks post conception and publicly available RNA expression data for four established and potential astroglial markers - GFAP, S100, AQP4 and YKL-40. In the hippocampal region, we also screened for C3, a complement component highly expressed in A1-reactive astrocytes. We found diverging partly overlapping patterns of the established astroglial markers GFAP, S100 and AQP4, confirming that none of these markers can fully describe and discriminate different developmental forms and subpopulations of astrocytes in human developing brain, although AQP4 seems to be the most sensitive and specific marker for the astroglial lineage at midgestation. AQP4 characterizes a brain-wide water transport system in cerebral cortex with regional differences in immunoreactivity at midgestation. AQP4 distinguishes a vast proportion of astrocytes and subpopulations of radial glial cells destined for the astroglial lineage, including astrocytes determined for the future glia limitans and apical truncated radial glial cells in ganglionic eminences, devoid of GFAP and S100. YKL-40 and C3d, previously found in reactive astrocytes, stain different subpopulations of astrocytes/astroglial progenitors in developing hippocampus at midgestation and may characterize specific subpopulations of 'developmental astrocytes'. Our results clearly reflect that lack of pan-astrocytic markers necessitates the consideration of time, region, context and aim when choosing appropriate astroglial markers.
Collapse
Affiliation(s)
- Camilla Bjørnbak Holst
- Faculty of Health and Medical Sciences, Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Copenhagen, Denmark.,Department of Radiation Biology, Department of Oncology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Beltoft Brøchner
- Faculty of Health and Medical Sciences, Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Kristoffer Vitting-Seerup
- Brain Tumor Biology, Danish Cancer Society Research Centre, Danish Cancer Society, Copenhagen, Denmark
| | - Kjeld Møllgård
- Faculty of Health and Medical Sciences, Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Meyer G, González-Arnay E, Moll U, Nemajerova A, Tissir F, González-Gómez M. Cajal-Retzius neurons are required for the development of the human hippocampal fissure. J Anat 2019; 235:569-589. [PMID: 30861578 DOI: 10.1111/joa.12947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2019] [Indexed: 01/14/2023] Open
Abstract
Cajal-Retzius neurons (CRN) are the main source of Reelin in the marginal zone of the developing neocortex and hippocampus (HC). They also express the transcription factor p73 and are complemented by later-appearing GABAergic Reelin+ interneurons. The human dorsal HC forms at gestational week 10 (GW10), when it develops a rudimentary Ammonic plate and incipient dentate migration, although the dorsal hippocampal fissure (HF) remains shallow and contains few CRN. The dorsal HC transforms into the indusium griseum (IG), concurrently with the rostro-caudal appearance of the corpus callosum, by GW14-17. Dorsal and ventral HC merge at the site of the former caudal hem, which is located at the level of the future atrium of the lateral ventricle and closely connected with the choroid plexus. The ventral HC forms at GW11 in the temporal lobe. The ventral HF is wide open at GW14-16 and densely populated by large numbers of CRNs. These are in intimate contact with the meninges and meningeal blood vessels, suggesting signalling through diverse pathways. At GW17, the fissure deepens and begins to fuse, although it is still marked by p73/Reelin+ CRNs. The p73KO mouse illustrates the importance of p73 in CRN for HF formation. In the mutant, Tbr1/Reelin+ CRNs are born in the hem but do not leave it and subsequently disappear, so that the mutant cortex and HC lack CRN from the onset of corticogenesis. The HF is absent, which leads to profound architectonic alterations of the HC. To determine which p73 isoform is important for HF formation, isoform-specific TAp73- and DeltaNp73-deficient embryonic and early postnatal mice were examined. In both mutants, the number of CRNs was reduced, but each of their phenotypes was much milder than in the global p73KO mutant missing both isoforms. In the TAp73KO mice, the HF of the dorsal HC failed to form, but was present in the ventral HC. In the DeltaNp73KO mice, the HC had a mild patterning defect along with a shorter HF. Complex interactions between both isoforms in CRNs may contribute to their crucial activity in the developing brain.
Collapse
Affiliation(s)
- Gundela Meyer
- Department of Basic Medical Sciences, University La Laguna, La Laguna, Spain
| | | | - Ute Moll
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Alice Nemajerova
- Department of Pathology, Stony Brook University, Stony Brook, NY, USA
| | - Fadel Tissir
- Developmental Neurobiology Group, Institute of NeuroScience, UCL Louvain, Brussels, Belgium
| | | |
Collapse
|
20
|
Anstötz M, Karsak M, Rune GM. Integrity of Cajal-Retzius cells in the reeler-mouse hippocampus. Hippocampus 2018; 29:550-565. [PMID: 30394609 DOI: 10.1002/hipo.23049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 10/10/2018] [Accepted: 10/16/2018] [Indexed: 01/01/2023]
Abstract
Cajal-Retzius (CR) cells are early-born glutamatergic neurons that are primarily known as the early main source of the signal protein Reelin. In the reeler mutant, the absence of Reelin causes severe defects in the radial migration of neurons, resulting in abnormal cortical layering. To date, the exact morphological properties of CR-cells independent of Reelin are unknown. With this in view, we studied the ontogenesis, density, and distribution of CR-cells in reeler mice that were cross-bred with a CXCR4-EGFP reporter mouse line, thus enabling us to clearly identify CR-cells positions in the disorganized hippocampus of the reeler mouse. As evidenced by morphological analysis, differences were found regarding CR-cell distribution and density: generally, we found fewer CR-cells in the developing and adult reeler hippocampus as compared to the hippocampus of wild-type animals (WT); however, in reeler mice, CR-cells were much more closely associated to the hippocampal fissure (HF), resulting in relatively higher local CR-cell densities. This higher local cell density was accompanied by stronger immunoreactivity of the CXCR4 ligand, stroma-derived factor-1 (SDF-1) that is known to regulate CR-cell positioning. Importantly, confocal microscopy indicates an integration of CR-cells into the developing and adult hippocampal network in reeler mice, raising evidence that network integration of CR-cells might be independent of Reelin.
Collapse
Affiliation(s)
- Max Anstötz
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Meliha Karsak
- Neuronal and Cellular Signal Transduction, Center for Molecular Neurobiology Hamburg (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
21
|
Watanabe K, Irie K, Hanashima C, Takebayashi H, Sato N. Diencephalic progenitors contribute to the posterior septum through rostral migration along the hippocampal axonal pathway. Sci Rep 2018; 8:11728. [PMID: 30082833 PMCID: PMC6078977 DOI: 10.1038/s41598-018-30020-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/23/2018] [Indexed: 12/21/2022] Open
Abstract
Septal nuclei are telencephalic structures associated with a variety of brain functions as part of the limbic system. The two posterior septal nuclei, the triangular septal nucleus (TS) and the bed nuclei of the anterior commissure (BAC), are involved in fear and anxiety through their projections to the medial habenular nucleus. However, the development of both the TS and BAC remains unclear. Here, we found a novel caudal origin and putative migratory stream of mouse posterior septal neurons arising from the thalamic eminence (TE), a transient developmental structure at the rostral end of the rodent diencephalon. TE-derived cells, which have glutamatergic identity, migrated rostrally and entered the telencephalic territory by passing beneath the third ventricle. Subsequently, they turned dorsally toward the posterior septum. We also observed that TS and BAC neurons in the postnatal septum were labeled with GFP by in utero electroporation into the TE, suggesting a shared origin. Furthermore, TE-derived septal neurons migrated along the fornix, an efferent pathway from the hippocampus. These results demonstrate that posterior septal neurons have a distinct extratelencephalic origin from other septal nuclei. This heterogeneous origin may contribute to neuronal diversity of the septal nuclear complex.
Collapse
Affiliation(s)
- Keisuke Watanabe
- Division of Gross Anatomy and Morphogenesis, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan.
| | - Koichiro Irie
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan.,Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| | - Noboru Sato
- Division of Gross Anatomy and Morphogenesis, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8510, Japan
| |
Collapse
|
22
|
Meyer G, González-Gómez M. The heterogeneity of human Cajal-Retzius neurons. Semin Cell Dev Biol 2018; 76:101-111. [DOI: 10.1016/j.semcdb.2017.08.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/24/2017] [Accepted: 08/28/2017] [Indexed: 12/29/2022]
|
23
|
Ruiz-Reig N, Andrés B, Huilgol D, Grove EA, Tissir F, Tole S, Theil T, Herrera E, Fairén A. Lateral Thalamic Eminence: A Novel Origin for mGluR1/Lot Cells. Cereb Cortex 2018; 27:2841-2856. [PMID: 27178193 DOI: 10.1093/cercor/bhw126] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A unique population of cells, called "lot cells," circumscribes the path of the lateral olfactory tract (LOT) in the rodent brain and acts to restrict its position at the lateral margin of the telencephalon. Lot cells were believed to originate in the dorsal pallium (DP). We show that Lhx2 null mice that lack a DP show a significant increase in the number of mGluR1/lot cells in the piriform cortex, indicating a non-DP origin of these cells. Since lot cells present common developmental features with Cajal-Retzius (CR) cells, we analyzed Wnt3a- and Dbx1-reporter mouse lines and found that mGluR1/lot cells are not generated in the cortical hem, ventral pallium, or septum, the best characterized sources of CR cells. Finally, we identified a novel origin for the lot cells by combining in utero electroporation assays and histochemical characterization. We show that mGluR1/lot cells are specifically generated in the lateral thalamic eminence and that they express mitral cell markers, although a minority of them express ΔNp73 instead. We conclude that most mGluR1/lot cells are prospective mitral cells migrating to the accessory olfactory bulb (OB), whereas mGluR1+, ΔNp73+ cells are CR cells that migrate through the LOT to the piriform cortex and the OB.
Collapse
Affiliation(s)
- Nuria Ruiz-Reig
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández, CSIC - UMH), San Juan de Alicante, Spain
| | - Belén Andrés
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández, CSIC - UMH), San Juan de Alicante, Spain
| | - Dhananjay Huilgol
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.,Current address: Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Fadel Tissir
- Université catholique de Louvain, Institute of Neuroscience, Brussels, Belgium
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Thomas Theil
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Eloisa Herrera
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández, CSIC - UMH), San Juan de Alicante, Spain
| | - Alfonso Fairén
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas - Universidad Miguel Hernández, CSIC - UMH), San Juan de Alicante, Spain
| |
Collapse
|
24
|
Kask K, Tikker L, Ruisu K, Lulla S, Oja EM, Meier R, Raid R, Velling T, Tõnissoo T, Pooga M. Targeted deletion of RIC8A in mouse neural precursor cells interferes with the development of the brain, eyes, and muscles. Dev Neurobiol 2018; 78:374-390. [PMID: 29380551 DOI: 10.1002/dneu.22578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 11/11/2022]
Abstract
Autosomal recessive disorders such as Fukuyama congenital muscular dystrophy, Walker-Warburg syndrome, and the muscle-eye-brain disease are characterized by defects in the development of patient's brain, eyes, and skeletal muscles. These syndromes are accompanied by brain malformations like type II lissencephaly in the cerebral cortex with characteristic overmigrations of neurons through the breaches of the pial basement membrane. The signaling pathways activated by laminin receptors, dystroglycan and integrins, control the integrity of the basement membrane, and their malfunctioning may underlie the pathologies found in the rise of defects reminiscent of these syndromes. Similar defects in corticogenesis and neuromuscular disorders were found in mice when RIC8A was specifically removed from neural precursor cells. RIC8A regulates a subset of G-protein α subunits and in several model organisms, it has been reported to participate in the control of cell division, signaling, and migration. Here, we studied the role of RIC8A in the development of the brain, muscles, and eyes of the neural precursor-specific conditional Ric8a knockout mice. The absence of RIC8A severely affected the attachment and positioning of radial glial processes, Cajal-Retzius' cells, and the arachnoid trabeculae, and these mice displayed additional defects in the lens, skeletal muscles, and heart development. All the discovered defects might be linked to aberrancies in cell adhesion and migration, suggesting that RIC8A has a crucial role in the regulation of cell-extracellular matrix interactions and that its removal leads to the phenotype characteristic to type II lissencephaly-associated diseases. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 374-390, 2018.
Collapse
Affiliation(s)
- Keiu Kask
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Laura Tikker
- Department of Biosciences, University of Helsinki, P.O. Box 56, Viikinkaari 9, FIN-00014, Helsinki, Finland
| | - Katrin Ruisu
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Sirje Lulla
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Eva-Maria Oja
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Riho Meier
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Raivo Raid
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Teet Velling
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Tambet Tõnissoo
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia
| | - Margus Pooga
- Institute of Molecular and Cell Biology, University of Tartu, 23 Riia St, Tartu, 51010, Estonia.,Institute of Technology, University of Tartu, Nooruse 1, Tartu, 50411, Estonia
| |
Collapse
|
25
|
Charting the protomap of the human telencephalon. Semin Cell Dev Biol 2017; 76:3-14. [PMID: 28834762 DOI: 10.1016/j.semcdb.2017.08.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/15/2017] [Indexed: 12/16/2022]
Abstract
The cerebral cortex is divided stereotypically into a number of functionally distinct areas. According to the protomap hypothesis formulated by Rakic neural progenitors in the ventricular zone form a mosaic of proliferative units that provide a primordial species-specific cortical map. Positional information of newborn neurons is maintained during their migration to the overlying cortical plate. Much evidence has been found to support this hypothesis from studies of primary cortical areas in mouse models in particular. Differential expansion of cortical areas and the introduction of new functional modules during evolution might be the result of changes in the progenitor cells. The human cerebral cortex shows a wide divergence from the mouse containing a much higher proportion of association cortex and a more complicated regionalised repertoire of neuron sub-types. To what extent does the protomap hypothesis hold true for the primate brain? This review summarises a growing number of studies exploring arealised gene expression in the early developing human telencephalon. The evidence so far is that the human and mouse brain do share fundamental mechanisms of areal specification, however there are subtle differences which could lead us to a better understanding of cortical evolution and the origins of neurodevelopmental diseases.
Collapse
|
26
|
Shah B, Püschel AW. Regulation of Rap GTPases in mammalian neurons. Biol Chem 2017; 397:1055-69. [PMID: 27186679 DOI: 10.1515/hsz-2016-0165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Abstract
Small GTPases are central regulators of many cellular processes. The highly conserved Rap GTPases perform essential functions in the mammalian nervous system during development and in mature neurons. During neocortical development, Rap1 is required to regulate cadherin- and integrin-mediated adhesion. In the adult nervous system Rap1 and Rap2 regulate the maturation and plasticity of dendritic spine and synapses. Although genetic studies have revealed important roles of Rap GTPases in neurons, their regulation by guanine nucleotide exchange factors (GEFs) that activate them and GTPase activating proteins (GAPs) that inactivate them by stimulating their intrinsic GTPase activity is just beginning to be explored in vivo. Here we review how GEFs and GAPs regulate Rap GTPases in the nervous system with a focus on their in vivo function.
Collapse
|
27
|
Meyer G, González-Gómez M. The Subpial Granular Layer and Transient Versus Persisting Cajal-Retzius Neurons of the Fetal Human Cortex. Cereb Cortex 2017; 28:2043-2058. [DOI: 10.1093/cercor/bhx110] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/18/2017] [Indexed: 12/20/2022] Open
Affiliation(s)
- Gundela Meyer
- Units of Anatomy (MGG) and Histology (GM), Department of Basic Medical Science, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| | - Miriam González-Gómez
- Units of Anatomy (MGG) and Histology (GM), Department of Basic Medical Science, Faculty of Medicine, University of La Laguna, Tenerife, Spain
| |
Collapse
|
28
|
Watanabe M, Fung ES, Chan FB, Wong JS, Coutts M, Monuki ES. BMP4 acts as a dorsal telencephalic morphogen in a mouse embryonic stem cell culture system. Biol Open 2016; 5:1834-1843. [PMID: 27815243 PMCID: PMC5200901 DOI: 10.1242/bio.012021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 10/28/2016] [Indexed: 12/25/2022] Open
Abstract
The concept of a morphogen - a molecule that specifies two or more cell fates in a concentration-dependent manner - is paradigmatic in developmental biology. Much remains unknown, however, about the existence of morphogens in the developing vertebrate central nervous system (CNS), including the mouse dorsal telencephalic midline (DTM). Bone morphogenetic proteins (BMPs) are candidate DTM morphogens, and our previous work demonstrated BMP4 sufficiency to induce one DTM cell fate - that of choroid plexus epithelial cells (CPECs) - in a mouse embryonic stem cell (mESC) culture system. Here we used BMP4 in a modified mESC culture system to derive a second DTM fate, the cortical hem (CH). CH and CPEC markers were induced by BMP4 in a concentration-dependent manner consistent with in vivo development. BMP4 concentrations that led to CH fate also promoted markers for Cajal-Retzius neurons, which are known CH derivatives. Interestingly, single BMP4 administrations also sufficed for appropriate temporal regulation of CH, CPEC, and cortical genes, with initially broad and overlapping dose-response profiles that sharpened over time. BMP4 concentrations that yielded CH- or CPEC-enriched populations also had different steady-state levels of phospho-SMAD1/5/8, suggesting that differences in BMP signaling intensity underlie DTM fate choice. Surprisingly, inactivation of the cortical selector gene Lhx2 did not affect DTM expression levels, dose-response profiles, or timing in response to BMP4, although neural progenitor genes were downregulated. These data indicate that BMP4 can act as a classic morphogen to orchestrate both spatial and temporal aspects of DTM fate acquisition, and can do so in the absence of Lhx2.
Collapse
Affiliation(s)
- Momoko Watanabe
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697-2300, USA
| | - Ernest S Fung
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Felicia B Chan
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Jessica S Wong
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Margaret Coutts
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Edwin S Monuki
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697-2300, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697-1705, USA
| |
Collapse
|
29
|
Castañeyra-Ruiz L, González-Marrero I, Hernández-Abad LG, Carmona-Calero EM, Meyer G, Castañeyra-Perdomo A. A Distal to Proximal Gradient of Human Choroid Plexus Development, with Antagonistic Expression of Glut1 and AQP1 in Mature Cells vs. Calbindin and PCNA in Proliferative Cells. Front Neuroanat 2016; 10:87. [PMID: 27721744 PMCID: PMC5034118 DOI: 10.3389/fnana.2016.00087] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/08/2016] [Indexed: 12/27/2022] Open
Abstract
The choroid plexuses (ChP) are highly vascularized tissues suspended from each of the cerebral ventricles. Their main function is to secret cerebrospinal fluid (CSF) that fills the ventricles and the subarachnoid spaces, forming a crucial system for the development and maintenance of the CNS. However, despite the essential role of the ChP–CSF system to regulate the CNS in a global manner, it still remains one of the most understudied areas in neurobiology. Here we define by immunohistochemistry the expression of different proteins involved in the maturation and functionality of the ChP from the late embryological period to maturity. We found an opposite gradient of expression between aquaporin 1 (AQP1) and glucose transporter 1 (Glut 1) that define functional maturation in the ChP periphery, and proliferating cell nuclear antigen (PCNA) and calbindin (CB), present in the ChP root zone with proliferative activity. We conclude that the maturation of the ChP matures from distal to proximal, starting in the areas nearest to the cortex, expressing in the distal, mature areas AQP1 and Glut1 (related to ChP functionality to support cortex development), and in the proximal immature areas (ChP root) CB and PCNA related to progenitor activity and proliferation.
Collapse
Affiliation(s)
- Leandro Castañeyra-Ruiz
- Department of Neurosurgery, School of Medicine, Washington University in Saint LouisSt. Louis, MO, USA; Unidad de Anatomía, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La LagunaLa Laguna, Spain
| | - Ibrahim González-Marrero
- Unidad de Anatomía, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna La Laguna, Spain
| | - Luis G Hernández-Abad
- Instituto de Investigación y Ciencia de Puerto de Rosario (INIPRO) Fuerteventura, Spain
| | - Emilia M Carmona-Calero
- Unidad de Anatomía, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna La Laguna, Spain
| | - Gundela Meyer
- Unidad de Anatomía, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La LagunaLa Laguna, Spain; Instituto de Investigación y Ciencia de Puerto de Rosario (INIPRO)Fuerteventura, Spain
| | - Agustín Castañeyra-Perdomo
- Unidad de Anatomía, Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La LagunaLa Laguna, Spain; Instituto de Investigación y Ciencia de Puerto de Rosario (INIPRO)Fuerteventura, Spain
| |
Collapse
|
30
|
Cell Type-Specific Circuit Mapping Reveals the Presynaptic Connectivity of Developing Cortical Circuits. J Neurosci 2016; 36:3378-90. [PMID: 26985044 DOI: 10.1523/jneurosci.0375-15.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED The mammalian cerebral cortex is a dense network composed of local, subcortical, and intercortical synaptic connections. As a result, mapping cell type-specific neuronal connectivity in the cerebral cortex in vivo has long been a challenge for neurobiologists. In particular, the development of excitatory and inhibitory interneuron presynaptic input has been hard to capture. We set out to analyze the development of this connectivity in the first postnatal month using a murine model. First, we surveyed the connectivity of one of the earliest populations of neurons in the brain, the Cajal-Retzius (CR) cells in the neocortex, which are known to be critical for cortical layer formation and are hypothesized to be important in the establishment of early cortical networks. We found that CR cells receive inputs from deeper-layer excitatory neurons and inhibitory interneurons in the first postnatal week. We also found that both excitatory pyramidal neurons and inhibitory interneurons received broad inputs in the first postnatal week, including inputs from CR cells. Expanding our analysis into the more mature brain, we assessed the inputs onto inhibitory interneurons and excitatory projection neurons, labeling neuronal progenitors with Cre drivers to study discrete populations of neurons in older cortex, and found that excitatory cortical and subcortical inputs are refined by the fourth week of development, whereas local inhibitory inputs increase during this postnatal period. Cell type-specific circuit mapping is specific, reliable, and effective, and can be used on molecularly defined subtypes to determine connectivity in the cortex. SIGNIFICANCE STATEMENT Mapping cortical connectivity in the developing mammalian brain has been an intractable problem, in part because it has not been possible to analyze connectivity with cell subtype precision. Our study systematically targets the presynaptic connections of discrete neuronal subtypes in both the mature and developing cerebral cortex. We analyzed the connections that Cajal-Retzius cells make and receive, and found that these cells receive inputs from deeper-layer excitatory neurons and inhibitory interneurons in the first postnatal week. We assessed the inputs onto inhibitory interneurons and excitatory projection neurons, the major two types of neurons in the cortex, and found that excitatory inputs are refined by the fourth week of development, whereas local inhibitory inputs increase during this postnatal period.
Collapse
|
31
|
Gabbott PLA. "Subpial Fan Cell" - A Class of Calretinin Neuron in Layer 1 of Adult Monkey Prefrontal Cortex. Front Neuroanat 2016; 10:28. [PMID: 27147978 PMCID: PMC4829592 DOI: 10.3389/fnana.2016.00028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
Layer 1 of the cortex contains populations of neurochemically distinct neurons and afferent fibers which markedly affect neural activity in the apical dendritic tufts of pyramidal cells. Understanding the causal mechanisms requires knowledge of the cellular architecture and synaptic organization of layer 1. This study has identified eight morphological classes of calretinin immunopositive (CRet+) neurons (including Cajal-Retzius cells) in layer 1 of the prefrontal cortex (PFC) in adult monkey (Macaca fasicularis), with a distinct class — termed “subpial fan (SPF) cell” — described in detail. SPF cells were rare horizontal unipolar CRet+ cells located directly beneath the pia with a single thick primary dendrite that branched into a characteristic fan-like dendritic tree tangential to the pial surface. Dendrites had spines, filamentous processes and thorny branchlets. SPF cells lay millimeters apart with intralaminar axons that ramified widely in upper layer 1. Such cells were GABA immunonegative (-) and occurred in areas beyond PFC. Interspersed amidst SPF cells displaying normal structural integrity were degenerating CRet+ neurons (including SPF cells) and clumps of lipofuscin-rich cellular debris. The number of degenerating SPF cells increased during adulthood. Ultrastructural analyses indicated SPF cell somata received asymmetric (A — presumed excitatory) and symmetric (S — presumed inhibitory) synaptic contacts. Proximal dendritic shafts received mainly S-type and distal shafts mostly A-type input. All dendritic thorns and most dendritic spines received both synapse types. The tangential areal density of SPF cell axonal varicosities varied radially from parent somata — with dense clusters in more distal zones. All boutons formed A-type contacts with CRet- structures. The main post-synaptic targets were dendritic shafts (67%; mostly spine-bearing) and dendritic spines (24%). SPF-SPF cell innervation was not observed. Morphometry of SPF cells indicated a unique class of CRet+/GABA- neuron in adult monkey PFC — possibly a subtype of persisting Cajal-Retzius cell. The distribution and connectivity of SPF cells suggest they act as integrative hubs in upper layer 1 during postnatal maturation. The main synaptic output of SPF cells likely provides a transminicolumnar excitatory influence across swathes of apical dendritic tufts — thus affecting information processing in discrete patches of layer 1 in adult monkey PFC.
Collapse
Affiliation(s)
- Paul L A Gabbott
- Neural Architectonics CentreOxford, UK; Department of Life, Health, and Chemical Sciences, The Open UniversityMilton Keynes, UK; University Department of Pharmacology, University of OxfordOxford, UK
| |
Collapse
|
32
|
Tkachenko LA, Zykin PA, Nasyrov RA, Krasnoshchekova EI. Distinctive Features of the Human Marginal Zone and Cajal-Retzius Cells: Comparison of Morphological and Immunocytochemical Features at Midgestation. Front Neuroanat 2016; 10:26. [PMID: 27047346 PMCID: PMC4797683 DOI: 10.3389/fnana.2016.00026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/04/2016] [Indexed: 12/13/2022] Open
Abstract
Despite a long history of research of cortical marginal zone (MZ) organization and development, a number of issues remain unresolved. One particular issue is the problem of Cajal-Retzius cells (C-R) identification. It is currently based on morphology and Reelin expression. The aim of this research is to investigate MZ cytoarchitectonics and Reelin-producing cells morphotypes in the superior temporal, pre- and postcentral cortex at GW24-26. We used Reelin (Reln) as the marker for C-R cells and microtubule-associated protein 2 (MAP2) and neurofilament heavy chain protein (N200) as markers of neuronal maturation. The MZ of all of the investigated areas had the distinct cytoarchitectonic of alternating cell sparse (MZP, SR) and cell dense (SGL, DGL) layers. The distribution of the neuromarkers across the MZ also showed layer specificity. MAP2-positive cells were only found in the SGL. N200 and Reelin-positive neurons in the MZP. N200-positive processes were forming a plexus at the DGL level. All of the N200-positive neurons found were in the MZP and had distinctive morphological features of C-R cells. All of the N200-positive neurons in MZ were also positive for Reelin, whereas MAP2-positive cells lack Reelin. Thus, the joint use of two immunomarkers allowed us to discern the C-R cells based on their morphotype and neurochemistry and indicate that the Reelin-positive cells of MZ at 24-26 GW were morphologically C-R cells. In the current study, we identified three C-R cells morphotypes. Using a 3D reconstruction, we made sure that all of them belonged to the single morphotype of triangular C-R cells. This approach will allow future studies to separate C-R cells from other Reelin-producing neurons which appear at later corticogenesis stages. In addition, our findings support the assumption that a plexus could be formed not only with C-R cells processes but also possibly by other cell processes by the poorly researched DGL, which is only allocated as a part of the human MZ.
Collapse
Affiliation(s)
- Lyubov A. Tkachenko
- Laboratory of Functional Neuromorphology, Department of Cytology and Histology, Saint-Petersburg State UniversitySaint-Petersburg, Russia
| | - Pavel A. Zykin
- Laboratory of Functional Neuromorphology, Department of Cytology and Histology, Saint-Petersburg State UniversitySaint-Petersburg, Russia
| | - Ruslan A. Nasyrov
- Department of Pathological Anatomy, Saint-Petersburg State Pediatric Medical UniversitySaint-Petersburg, Russia
| | - Elena I. Krasnoshchekova
- Laboratory of Functional Neuromorphology, Department of Cytology and Histology, Saint-Petersburg State UniversitySaint-Petersburg, Russia
| |
Collapse
|
33
|
Barber M, Pierani A. Tangential migration of glutamatergic neurons and cortical patterning during development: Lessons from Cajal-Retzius cells. Dev Neurobiol 2015; 76:847-81. [PMID: 26581033 DOI: 10.1002/dneu.22363] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 11/12/2015] [Accepted: 11/13/2015] [Indexed: 12/14/2022]
Abstract
Tangential migration is a mode of cell movement, which in the developing cerebral cortex, is defined by displacement parallel to the ventricular surface and orthogonal to the radial glial fibers. This mode of long-range migration is a strategy by which distinct neuronal classes generated from spatially and molecularly distinct origins can integrate to form appropriate neural circuits within the cortical plate. While it was previously believed that only GABAergic cortical interneurons migrate tangentially from their origins in the subpallial ganglionic eminences to integrate in the cortical plate, it is now known that transient populations of glutamatergic neurons also adopt this mode of migration. These include Cajal-Retzius cells (CRs), subplate neurons (SPs), and cortical plate transient neurons (CPTs), which have crucial roles in orchestrating the radial and tangential development of the embryonic cerebral cortex in a noncell-autonomous manner. While CRs have been extensively studied, it is only in the last decade that the molecular mechanisms governing their tangential migration have begun to be elucidated. To date, the mechanisms of SPs and CPTs tangential migration remain unknown. We therefore review the known signaling pathways, which regulate parameters of CRs migration including their motility, contact-redistribution and adhesion to the pial surface, and discuss this in the context of how CR migration may regulate their signaling activity in a spatial and temporal manner. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 847-881, 2016.
Collapse
Affiliation(s)
- Melissa Barber
- Institut Jacques-Monod, CNRS, Université Paris Diderot, Sorbonne Cité, Paris, France.,Department of Cell and Developmental Biology, University College London, WC1E 6BT, United Kingdom
| | - Alessandra Pierani
- Institut Jacques-Monod, CNRS, Université Paris Diderot, Sorbonne Cité, Paris, France
| |
Collapse
|
34
|
Montiel JF, Aboitiz F. Pallial patterning and the origin of the isocortex. Front Neurosci 2015; 9:377. [PMID: 26512233 PMCID: PMC4604247 DOI: 10.3389/fnins.2015.00377] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 09/28/2015] [Indexed: 12/30/2022] Open
Abstract
Together with a complex variety of behavioral, physiological, morphological, and neurobiological innovations, mammals are characterized by the development of an extensive isocortex (also called neocortex) that is both laminated and radially organized, as opposed to the brain of birds and reptiles. In this article, we will advance a developmental hypothesis in which the mechanisms of evolutionary brain growth remain partly conserved across amniotes (mammals, reptiles and birds), all based on Pax6 signaling or related morphogens. Despite this conservatism, only in mammals there is an additional upregulation of dorsal and anterior signaling centers (the cortical hem and the anterior forebrain, respectively) that promoted a laminar and a columnar structure into the neocortex. It is possible that independently, some birds also developed an upregulated dorsal pallium.
Collapse
Affiliation(s)
- Juan F. Montiel
- Facultad de Medicina, Centro de Investigación Biomédica, Universidad Diego PortalesSantiago, Chile
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of OxfordOxford, UK
| | - Francisco Aboitiz
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de ChileSantiago, Chile
| |
Collapse
|
35
|
Adutwum-Ofosu KK, Magnani D, Theil T, Price DJ, Fotaki V. The molecular and cellular signatures of the mouse eminentia thalami support its role as a signalling centre in the developing forebrain. Brain Struct Funct 2015; 221:3709-27. [PMID: 26459142 PMCID: PMC5009181 DOI: 10.1007/s00429-015-1127-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/30/2015] [Indexed: 12/20/2022]
Abstract
The mammalian eminentia thalami (EmT) (or thalamic eminence) is an embryonic forebrain structure of unknown function. Here, we examined the molecular and cellular properties of the mouse EmT. We first studied mRNA expression of signalling molecules and found that the EmT is a structure, rich in expression of secreted factors, with Wnts being the most abundantly detected. We then examined whether EmT tissue could induce cell fate changes when grafted ectopically. For this, we transplanted EmT tissue from a tau-GFP mouse to the ventral telencephalon of a wild type host, a telencephalic region where Wnt signalling is not normally active but which we showed in culture experiments is competent to respond to Wnts. We observed that the EmT was able to induce in adjacent ventral telencephalic cells ectopic expression of Lef1, a transcriptional activator and a target gene of the Wnt/β-catenin pathway. These Lef1-positive;GFP-negative cells expressed the telencephalic marker Foxg1 but not Ascl1, which is normally expressed by ventral telencephalic cells. These results suggest that the EmT has the capacity to activate Wnt/β-catenin signalling in the ventral telencephalon and to suppress ventral telencephalic gene expression. Altogether, our data support a role of the EmT as a signalling centre in the developing mouse forebrain.
Collapse
Affiliation(s)
- Kevin Kofi Adutwum-Ofosu
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,Department of Anatomy, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Dario Magnani
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.,MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Thomas Theil
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - David J Price
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Vassiliki Fotaki
- The University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
36
|
Squarzoni P, Thion MS, Garel S. Neuronal and microglial regulators of cortical wiring: usual and novel guideposts. Front Neurosci 2015; 9:248. [PMID: 26236185 PMCID: PMC4505395 DOI: 10.3389/fnins.2015.00248] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/30/2015] [Indexed: 12/17/2022] Open
Abstract
Neocortex functioning relies on the formation of complex networks that begin to be assembled during embryogenesis by highly stereotyped processes of cell migration and axonal navigation. The guidance of cells and axons is driven by extracellular cues, released along by final targets or intermediate targets located along specific pathways. In particular, guidepost cells, originally described in the grasshopper, are considered discrete, specialized cell populations located at crucial decision points along axonal trajectories that regulate tract formation. These cells are usually early-born, transient and act at short-range or via cell-cell contact. The vast majority of guidepost cells initially identified were glial cells, which play a role in the formation of important axonal tracts in the forebrain, such as the corpus callosum, anterior, and post-optic commissures as well as optic chiasm. In the last decades, tangential migrating neurons have also been found to participate in the guidance of principal axonal tracts in the forebrain. This is the case for several examples such as guideposts for the lateral olfactory tract (LOT), corridor cells, which open an internal path for thalamo-cortical axons and Cajal-Retzius cells that have been involved in the formation of the entorhino-hippocampal connections. More recently, microglia, the resident macrophages of the brain, were specifically observed at the crossroads of important neuronal migratory routes and axonal tract pathways during forebrain development. We furthermore found that microglia participate to the shaping of prenatal forebrain circuits, thereby opening novel perspectives on forebrain development and wiring. Here we will review the last findings on already known guidepost cell populations and will discuss the role of microglia as a potentially new class of atypical guidepost cells.
Collapse
Affiliation(s)
- Paola Squarzoni
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Morgane S Thion
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| | - Sonia Garel
- Centre National de la Recherche Scientifique UMR8197, Ecole Normale Supérieure, Institut de Biologie, Institut National de la Santé et de la Recherche Médicale U1024 Paris, France
| |
Collapse
|
37
|
Skorput AGJ, Yeh HH. Effects of ethanol exposure in utero on Cajal-Retzius cells in the developing cortex. Alcohol Clin Exp Res 2015; 39:853-62. [PMID: 25845402 DOI: 10.1111/acer.12696] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/31/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Prenatal exposure to ethanol exerts teratogenic effects on the developing brain. Here, we tested the hypothesis that exposure to ethanol in utero alters the disposition of Cajal-Retzius cells that play a key role in orchestrating proliferation, migration, and laminar integration of cortical neurons in the embryonic cortex. METHODS Pregnant Ebf2-EGFP mice, harboring EGFP-fluorescent Cajal-Retzius cells, were subjected to a 2% w/w ethanol consumption regimen starting at neural tube closure and lasting throughout gestation. Genesis of Cajal-Retzius cells was assessed by means of 5-bromo-2-deoxyuridine (BrdU) immunofluorescence at embryonic day 12.5, their counts and distribution were determined between postnatal day (P)0 and P4, patch clamp electrophysiology was performed between P2 and P3 to analyze GABA-mediated synaptic activity, and open-field behavioral testing was conducted in P45-P50 adolescents. RESULTS In Ebf2-EGFP embryos exposed to ethanol in utero, we found increased BrdU labeling and expanded distribution of Cajal-Retzius cells in the cortical hem, pointing to increased genesis and proliferation. Postnatally, we found an increase in Cajal-Retzius cell number in cortical layer I. In addition, they displayed altered patterning of spontaneous GABA-mediated synaptic barrages and enhanced GABA-mediated synaptic activity, suggesting enhanced GABAergic tone. CONCLUSIONS These findings, together, underscore that Cajal-Retzius cells contribute to the ethanol-induced aberration of cortical development and abnormal GABAergic neurotransmission at the impactful time when intracortical circuits form.
Collapse
Affiliation(s)
- Alexander G J Skorput
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | |
Collapse
|
38
|
Squier W, Jansen A. Polymicrogyria: pathology, fetal origins and mechanisms. Acta Neuropathol Commun 2014; 2:80. [PMID: 25047116 PMCID: PMC4149230 DOI: 10.1186/s40478-014-0080-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 01/28/2023] Open
Abstract
Polymicrogyria (PMG) is a complex cortical malformation which has so far defied any mechanistic or genetic explanation. Adopting a broad definition of an abnormally folded or festooned cerebral cortical neuronal ribbon, this review addresses the literature on PMG and the mechanisms of its development, as derived from the neuropathological study of many cases of human PMG, a large proportion in fetal life. This reveals the several processes which appear to be involved in the early stages of formation of polymicrogyric cortex. The most consistent feature of developing PMG is disruption of the brain surface with pial defects, over-migration of cells, thickening and reduplication of the pial collagen layers and increased leptomeningeal vascularity. Evidence from animal models is consistent with our observations and supports the notion that disturbance in the formation of the leptomeninges or loss of their normal signalling functions are potent contributors to cortical malformation. Other mechanisms which may lead to PMG include premature folding of the neuronal band, abnormal fusion of adjacent gyri and laminar necrosis of the developing cortex. The observation of PMG in association with other and better understood forms of brain malformation, such as cobblestone cortex, suggests mechanistic pathways for some forms of PMG. The role of altered physical properties of the thickened leptomeninges in exerting mechanical constraints on the developing cortex is also considered.
Collapse
|
39
|
Kumamoto T, Hanashima C. Neuronal subtype specification in establishing mammalian neocortical circuits. Neurosci Res 2014; 86:37-49. [PMID: 25019611 DOI: 10.1016/j.neures.2014.07.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 06/21/2014] [Accepted: 06/23/2014] [Indexed: 11/28/2022]
Abstract
The functional integrity of the neocortical circuit relies on the precise production of diverse neuron populations and their assembly during development. In recent years, extensive progress has been made in the understanding of the mechanisms that control differentiation of each neuronal type within the neocortex. In this review, we address how the elaborate neocortical cytoarchitecture is established from a simple neuroepithelium based on recent studies examining the spatiotemporal mechanisms of neuronal subtype specification. We further discuss the critical events that underlie the conversion of the stem amniotes cerebrum to a mammalian-type neocortex, and extend these key findings in the light of mammalian evolution to understand how the neocortex in humans evolved from common ancestral mammals.
Collapse
Affiliation(s)
- Takuma Kumamoto
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan
| | - Carina Hanashima
- Laboratory for Neocortical Development, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan; Department of Biology, Graduate School of Science, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
40
|
Arai Y, Pierani A. Development and evolution of cortical fields. Neurosci Res 2014; 86:66-76. [PMID: 24983875 DOI: 10.1016/j.neures.2014.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Revised: 06/05/2014] [Accepted: 06/10/2014] [Indexed: 11/17/2022]
Abstract
The neocortex is the brain structure that has been subjected to a major size expansion, in its relative size, during mammalian evolution. It arises from the cortical primordium through coordinated growth of neural progenitor cells along both the tangential and radial axes and their patterning providing spatial coordinates. Functional neocortical areas are ultimately consolidated by environmental influences such as peripheral sensory inputs. Throughout neocortical evolution, cortical areas have become more sophisticated and numerous. This increase in number is possibly involved in the complexification of neocortical function in primates. Whereas extensive divergence of functional cortical fields is observed during evolution, the fundamental mechanisms supporting the allocation of cortical areas and their wiring are conserved, suggesting the presence of core genetic mechanisms operating in different species. We will discuss some of the basic molecular mechanisms including morphogen-dependent ones involved in the precise orchestration of neurogenesis in different cortical areas, elucidated from studies in rodents. Attention will be paid to the role of Cajal-Retzius neurons, which were recently proposed to be migrating signaling units also involved in arealization, will be addressed. We will further review recent works on molecular mechanisms of cortical patterning resulting from comparative analyses between different species during evolution.
Collapse
Affiliation(s)
- Yoko Arai
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France.
| | - Alessandra Pierani
- Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France
| |
Collapse
|
41
|
Martínez-Cerdeño V, Noctor SC. Cajal, Retzius, and Cajal-Retzius cells. Front Neuroanat 2014; 8:48. [PMID: 24987337 PMCID: PMC4060955 DOI: 10.3389/fnana.2014.00048] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 05/29/2014] [Indexed: 01/06/2023] Open
Abstract
The marginal zone (MZ) of the prenatal cerebral cortex plays a crucial role in cellular migration and laminar patterning in the developing neocortex and its equivalent in the adult brain – layer I, participates in cortical circuitry integration within the adult neocortex. The MZ/layer I, which has also been called the plexiform layer and cell-poor zone of Meynert, among others, is home to several cell populations including glia, neurons, and Cajal–Retzius (CR) cells. Cajal once said that the MZ is one of the oldest formations in the phylogenetic series, and that the characteristics of layer I in human are similar in all vertebrates except fish (Ramon y Cajal, 1899). Despite the presence of CR cells in the MZ/layer I of all developing and adult vertebrate brains, and more than one hundred years of research, the phenotype and function of layer I cells have still not been clearly defined. Recent technological advances have yielded significant progress in functional and developmental studies, but much remains to be understood about neurons in MZ/layer I. Since the time of Retzius and Cajal, and continuing with modern era research from the likes of Marín-Padilla, the study of CR cells has been based on their morphological characteristics in Golgi staining. However, since Cajal’s initial description, the term “CR cell” has been applied differently and now is often used to indicate reelin (Reln)-positive cells in MZ/layer I. Here we review the history of work by Cajal, Retzius, and others pertaining to CR cells. We will establish a link between original descriptions of CR cell morphology by Cajal, Retzius, and others, and current understandings of the cell populations that reside in MZ/layer I based on the use of cellular markers. We propose to use the term “CR cell” for the class of neurons that express Reln in the MZ/layer I in both prenatal, developing and adult cerebral cortex.
Collapse
Affiliation(s)
- Verónica Martínez-Cerdeño
- Institute for Pediatric Regenerative Medicine, University of California at Davis Sacramento, CA, USA ; Medical Pathology and Laboratory Medicine, University of California at Davis Sacramento, CA, USA ; MIND Institute, University of California at Davis Sacramento, CA, USA
| | - Stephen C Noctor
- MIND Institute, University of California at Davis Sacramento, CA, USA ; Department of Psychiatry and Behavioral Sciences, University of California at Davis Sacramento, CA, USA
| |
Collapse
|
42
|
Kirischuk S, Luhmann HJ, Kilb W. Cajal-Retzius cells: update on structural and functional properties of these mystic neurons that bridged the 20th century. Neuroscience 2014; 275:33-46. [PMID: 24931764 DOI: 10.1016/j.neuroscience.2014.06.009] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/03/2014] [Accepted: 06/03/2014] [Indexed: 02/02/2023]
Abstract
Cajal-Retzius cells (CRc) represent a mostly transient neuronal cell type localized in the uppermost layer of the developing neocortex. The observation that CRc are a major source of the extracellular matrix protein reelin, which is essential for the laminar development of the cerebral cortex, attracted the interest in this unique cell type. In this review we will (i) describe the morphological and molecular properties of neocortical CRc, with a special emphasize on the question which markers can be used to identify CRc, (ii) summarize reports that identified the different developmental origins of CRc, (iii) discuss the fate of CRc, including recent evidence for apoptotic cell death and a possible persistence of some CRc, (iv) provide a detailed description of the electrical membrane properties and transmitter receptors of CRc, and (v) address the role of CRc in early neuronal circuits and cortical development. Finally, we speculate whether CRc may provide a link between early network activity and the structural maturation of neocortical circuits.
Collapse
Affiliation(s)
- S Kirischuk
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany
| | - H J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany
| | - W Kilb
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, D-55128 Mainz, Germany.
| |
Collapse
|
43
|
González-Gómez M, Meyer G. Dynamic expression of calretinin in embryonic and early fetal human cortex. Front Neuroanat 2014; 8:41. [PMID: 24917793 PMCID: PMC4042362 DOI: 10.3389/fnana.2014.00041] [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: 04/28/2014] [Accepted: 05/16/2014] [Indexed: 02/04/2023] Open
Abstract
Calretinin (CR) is one of the earliest neurochemical markers in human corticogenesis. In embryos from Carnegie stages (CS) 17 to 23, calbindin (CB) and CR stain opposite poles of the incipient cortex suggesting early regionalization: CB marks the neuroepithelium of the medial boundary of the cortex with the choroid plexus (cortical hem). By contrast, CR is confined to the subventricular zone (SVZ) of the lateral and caudal ganglionic eminences at the pallial-subpallial boundary (PSB, or antihem), from where CR+/Tbr1- neurons migrate toward piriform cortex and amygdala as a component of the lateral cortical stream. At CS 19, columns of CR+ cells arise in the rostral cortex, and contribute at CS 20 to the “monolayer” of horizontal Tbr1+/CR+ and GAD+ cells in the preplate. At CS 21, the “pioneer cortical plate” appears as a radial aggregation of CR+/Tbr1+ neurons, which cover the entire future neocortex and extend the first corticofugal axons. CR expression in early human corticogenesis is thus not restricted to interneurons, but is also present in the first excitatory projection neurons of the cortex. At CS 21/22, the cortical plate is established following a lateral to medial gradient, when Tbr1+/CR- neurons settle within the pioneer cortical plate, and thus separate superficial and deep pioneer neurons. CR+ pioneer neurons disappear shortly after the formation of the cortical plate. Reelin+ Cajal-Retzius cells begin to express CR around CS21 (7/8 PCW). At CS 21–23, the CR+ SVZ at the PSB is the source of CR+ interneurons migrating into the cortical SVZ. In turn, CB+ interneurons migrate from the subpallium into the intermediate zone following the fibers of the internal capsule. Early CR+ and CB+ interneurons thus have different origins and migratory routes. CR+ cell populations in the embryonic telencephalon take part in a complex sequence of events not analyzed so far in other mammalian species, which may represent a distinctive trait of the initial steps of human corticogenesis.
Collapse
Affiliation(s)
- Miriam González-Gómez
- Departamento de Anatomía, Facultad de Medicina, Universidad de La Laguna Tenerife, Spain
| | - Gundela Meyer
- Departamento de Anatomía, Facultad de Medicina, Universidad de La Laguna Tenerife, Spain
| |
Collapse
|
44
|
Gil V, Nocentini S, Del Río JA. Historical first descriptions of Cajal-Retzius cells: from pioneer studies to current knowledge. Front Neuroanat 2014; 8:32. [PMID: 24904301 PMCID: PMC4034043 DOI: 10.3389/fnana.2014.00032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/23/2014] [Indexed: 11/20/2022] Open
Abstract
Santiago Ramón y Cajal developed a great body of scientific research during the last decade of 19th century, mainly between 1888 and 1892, when he published more than 30 manuscripts. The neuronal theory, the structure of dendrites and spines, and fine microscopic descriptions of numerous neural circuits are among these studies. In addition, numerous cell types (neuronal and glial) were described by Ramón y Cajal during this time using this “reazione nera” or Golgi method. Among these neurons were the special cells of the molecular layer of the neocortex. These cells were also termed Cajal cells or Retzius cells by other colleagues. Today these cells are known as Cajal–Retzius cells. From the earliest description, several biological aspects of these fascinating cells have been analyzed (e.g., cell morphology, physiological properties, origin and cellular fate, putative function during cortical development, etc). In this review we will summarize in a temporal basis the emerging knowledge concerning this cell population with specific attention the pioneer studies of Santiago Ramón y Cajal.
Collapse
Affiliation(s)
- Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Parc Científic de Barcelona Barcelona, Spain ; Department of Cell Biology, Faculty of Biology, University of Barcelona Barcelona, Spain ; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas Barcelona, Spain
| | - Sara Nocentini
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Parc Científic de Barcelona Barcelona, Spain ; Department of Cell Biology, Faculty of Biology, University of Barcelona Barcelona, Spain ; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas Barcelona, Spain
| | - José A Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, Parc Científic de Barcelona Barcelona, Spain ; Department of Cell Biology, Faculty of Biology, University of Barcelona Barcelona, Spain ; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas Barcelona, Spain
| |
Collapse
|
45
|
Gutzmann A, Ergül N, Grossmann R, Schultz C, Wahle P, Engelhardt M. A period of structural plasticity at the axon initial segment in developing visual cortex. Front Neuroanat 2014; 8:11. [PMID: 24653680 PMCID: PMC3949221 DOI: 10.3389/fnana.2014.00011] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/20/2014] [Indexed: 01/09/2023] Open
Abstract
Cortical networks are shaped by sensory experience and are most susceptible to modifications during critical periods characterized by enhanced plasticity at the structural and functional level. A system particularly well-studied in this context is the mammalian visual system. Plasticity has been documented for the somatodendritic compartment of neurons in detail. A neuronal microdomain not yet studied in this context is the axon initial segment (AIS) located at the proximal axon segment. It is a specific electrogenic axonal domain and the site of action potential (AP) generation. Recent studies showed that structure and function of the AIS can be dynamically regulated. Here we hypothesize that the AIS shows a dynamic regulation during maturation of the visual cortex. We therefore analyzed AIS length development from embryonic day (E) 12.5 to adulthood in mice. A tri-phasic time course of AIS length remodeling during development was observed. AIS first appeared at E14.5 and increased in length throughout the postnatal period to a peak between postnatal day (P) 10 to P15 (eyes open P13–14). Then, AIS length was reduced significantly around the beginning of the critical period for ocular dominance plasticity (CP, P21). Shortest AIS were observed at the peak of the CP (P28), followed by a moderate elongation toward the end of the CP (P35). To test if the dynamic maturation of the AIS is influenced by eye opening (onset of activity), animals were deprived of visual input before and during the CP. Deprivation for 1 week prior to eye opening did not affect AIS length development. However, deprivation from P0 to 28 and P14 to 28 resulted in AIS length distribution similar to the peak at P15. In other words, deprivation from birth prevents the transient shortening of the AIS and maintains an immature AIS length. These results are the first to suggest a dynamic maturation of the AIS in cortical neurons and point to novel mechanisms in the development of neuronal excitability.
Collapse
Affiliation(s)
- Annika Gutzmann
- CBTM, Medical Faculty Mannheim, Institute of Neuroanatomy, Heidelberg University Heidelberg, Germany
| | - Nursah Ergül
- CBTM, Medical Faculty Mannheim, Institute of Neuroanatomy, Heidelberg University Heidelberg, Germany
| | - Rebecca Grossmann
- CBTM, Medical Faculty Mannheim, Institute of Neuroanatomy, Heidelberg University Heidelberg, Germany
| | - Christian Schultz
- CBTM, Medical Faculty Mannheim, Institute of Neuroanatomy, Heidelberg University Heidelberg, Germany
| | - Petra Wahle
- AG Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Germany
| | - Maren Engelhardt
- CBTM, Medical Faculty Mannheim, Institute of Neuroanatomy, Heidelberg University Heidelberg, Germany
| |
Collapse
|
46
|
Bjørnbak C, Brøchner CB, Larsen LA, Johansen JS, Møllgård K. Brain barriers and a subpopulation of astroglial progenitors of developing human forebrain are immunostained for the glycoprotein YKL-40. J Histochem Cytochem 2014; 62:369-88. [PMID: 24595665 DOI: 10.1369/0022155414528514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
YKL-40, a glycoprotein involved in cell differentiation, has been associated with neurodevelopmental disorders, angiogenesis, neuroinflammation and glioblastomas. We evaluated YKL-40 protein distribution in the early human forebrain using double-labeling immunofluorescence and immunohistochemistry. Immunoreactivity was detected in neuroepithelial cells, radial glial end feet, leptomeningeal cells and choroid plexus epithelial cells. The subpial marginal zone was YKL-40-positive, particularly in the hippocampus, from an early beginning stage in its development. Blood vessels in the intermediate and subventricular zones showed specific YKL-40 reactivity confined to pericytes. Furthermore, a population of YKL-40-positive, small, rounded cells was identified in the ventricular and subventricular zones. Real-time quantitative RT-PCR analysis showed strong YKL-40 mRNA expression in the leptomeninges and the choroid plexuses, and weaker expression in the telencephalic wall. Immunohistochemistry revealed a differential distribution of YKL-40 across the zones of the developing telencephalic wall. We show that YKL-40 is associated with sites of the brain barrier systems and propose that it is involved in controlling local angiogenesis and access of peripheral cells to the forebrain via secretion from leptomeningeal cells, choroid plexus epithelium and pericytes. Furthermore, we suggest that the small, rounded, YKL-40-positive cells represent a subpopulation of astroglial progenitors, and that YKL-40 could be involved in the differentiation of a particular astrocytic lineage.
Collapse
Affiliation(s)
- Camilla Bjørnbak
- Department of Cellular and Molecular Medicine (CB,CBB,LAL,KM), Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
47
|
Jovanov Milošević N, Judaš M, Aronica E, Kostovic I. Neural ECM in laminar organization and connectivity development in healthy and diseased human brain. PROGRESS IN BRAIN RESEARCH 2014; 214:159-78. [DOI: 10.1016/b978-0-444-63486-3.00007-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Aboitiz F, Zamorano F. Neural progenitors, patterning and ecology in neocortical origins. Front Neuroanat 2013; 7:38. [PMID: 24273496 PMCID: PMC3824149 DOI: 10.3389/fnana.2013.00038] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 10/21/2013] [Indexed: 01/13/2023] Open
Abstract
The anatomical organization of the mammalian neocortex stands out among vertebrates for its laminar and columnar arrangement, featuring vertically oriented, excitatory pyramidal neurons. The evolutionary origin of this structure is discussed here in relation to the brain organization of other amniotes, i.e., the sauropsids (reptiles and birds). Specifically, we address the developmental modifications that had to take place to generate the neocortex, and to what extent these modifications were shared by other amniote lineages or can be considered unique to mammals. In this article, we propose a hypothesis that combines the control of proliferation in neural progenitor pools with the specification of regional morphogenetic gradients, yielding different anatomical results by virtue of the differential modulation of these processes in each lineage. Thus, there is a highly conserved genetic and developmental battery that becomes modulated in different directions according to specific selective pressures. In the case of early mammals, ecological conditions like nocturnal habits and reproductive strategies are considered to have played a key role in the selection of the particular brain patterning mechanisms that led to the origin of the neocortex.
Collapse
Affiliation(s)
- Francisco Aboitiz
- Departamento de Psiquiatría, Facultad de Medicina y Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile Santiago, Chile
| | | |
Collapse
|
49
|
Gilmore EC, Walsh CA. Genetic causes of microcephaly and lessons for neuronal development. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2013; 2:461-78. [PMID: 24014418 PMCID: PMC3767923 DOI: 10.1002/wdev.89] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The study of human developmental microcephaly is providing important insights into brain development. It has become clear that developmental microcephalies are associated with abnormalities in cellular production, and that the pathophysiology of microcephaly provides remarkable insights into how the brain generates the proper number of neurons that determine brain size. Most of the genetic causes of 'primary' developmental microcephaly (i.e., not associated with other syndromic features) are associated with centrosomal abnormalities. In addition to other functions, centrosomal proteins control the mitotic spindle, which is essential for normal cell proliferation during mitosis. However, the brain is often uniquely affected when microcephaly genes are mutated implying special centrosomal-related functions in neuronal production. Although models explaining how this could occur have some compelling data, they are not without controversy. Interestingly, some of the microcephaly genes show evidence that they were targets of evolutionary selection in primates and human ancestors, suggesting potential evolutionary roles in controlling neuronal number and brain volume across species. Mutations in DNA repair pathway genes also lead to microcephaly. Double-stranded DNA breaks appear to be a prominent type of damage that needs to be repaired during brain development, yet why defects in DNA repair affect the brain preferentially and if DNA repair relates to centrosome function, are not clearly understood.
Collapse
Affiliation(s)
- Edward C Gilmore
- Division of Pediatric Neurology, Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | | |
Collapse
|
50
|
Post-transcriptional regulatory elements and spatiotemporal specification of neocortical stem cells and projection neurons. Neuroscience 2013; 248:499-528. [PMID: 23727006 DOI: 10.1016/j.neuroscience.2013.05.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/15/2013] [Accepted: 05/21/2013] [Indexed: 11/22/2022]
Abstract
The mature neocortex is a unique six-layered mammalian brain region. It is composed of morphologically and functionally distinct subpopulations of primary projection neurons that form complex circuits across the central nervous system. The precisely-timed generation of projection neurons from neural stem cells governs their differentiation, postmitotic specification, and signaling, and is critical for cognitive and sensorimotor ability. Developmental perturbations to the birthdate, location, and connectivity of neocortical neurons are observed in neurological and psychiatric disorders. These facts are highlighting the importance of the precise spatiotemporal development of the neocortex regulated by intricate transcriptional, but also complex post-transcriptional events. Indeed, mRNA transcripts undergo many post-transcriptional regulatory steps before the production of functional proteins, which specify neocortical neural stem cells and subpopulations of neocortical neurons. Therefore, particular attention is paid to the differential post-transcriptional regulation of key transcripts by RNA-binding proteins, including splicing, localization, stability, and translation. We also present a transcriptome screen of candidate molecules associated with post-transcriptional mRNA processing that are differentially expressed at key developmental time points across neocortical prenatal neurogenesis.
Collapse
|