1
|
Iskusnykh IY, Fattakhov N, Li Y, Bihannic L, Kirchner MK, Steshina EY, Northcott PA, Chizhikov VV. Lmx1a is a master regulator of the cortical hem. eLife 2023; 12:e84095. [PMID: 37725078 PMCID: PMC10508884 DOI: 10.7554/elife.84095] [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: 10/11/2022] [Accepted: 09/05/2023] [Indexed: 09/21/2023] Open
Abstract
Development of the nervous system depends on signaling centers - specialized cellular populations that produce secreted molecules to regulate neurogenesis in the neighboring neuroepithelium. In some cases, signaling center cells also differentiate to produce key types of neurons. The formation of a signaling center involves its induction, the maintenance of expression of its secreted molecules, and cell differentiation and migration events. How these distinct processes are coordinated during signaling center development remains unknown. By performing studies in mice, we show that Lmx1a acts as a master regulator to orchestrate the formation and function of the cortical hem (CH), a critical signaling center that controls hippocampus development. Lmx1a co-regulates CH induction, its Wnt signaling, and the differentiation and migration of CH-derived Cajal-Retzius neurons. Combining RNAseq, genetic, and rescue experiments, we identified major downstream genes that mediate distinct Lmx1a-dependent processes. Our work revealed that signaling centers in the mammalian brain employ master regulatory genes and established a framework for analyzing signaling center development.
Collapse
Affiliation(s)
- Igor Y Iskusnykh
- Department of Anatomy and Neurobiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Nikolai Fattakhov
- Department of Anatomy and Neurobiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Yiran Li
- Department of Developmental Neurobiology, St. Jude Children's Research HospitalMemphisUnited States
| | - Laure Bihannic
- Department of Developmental Neurobiology, St. Jude Children's Research HospitalMemphisUnited States
| | - Matthew K Kirchner
- Department of Anatomy and Neurobiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Ekaterina Y Steshina
- Department of Anatomy and Neurobiology, University of Tennessee Health Science CenterMemphisUnited States
| | - Paul A Northcott
- Department of Developmental Neurobiology, St. Jude Children's Research HospitalMemphisUnited States
| | - Victor V Chizhikov
- Department of Anatomy and Neurobiology, University of Tennessee Health Science CenterMemphisUnited States
| |
Collapse
|
2
|
Moreau MX, Saillour Y, Elorriaga V, Bouloudi B, Delberghe E, Deutsch Guerrero T, Ochandorena-Saa A, Maeso-Alonso L, Marques MM, Marin MC, Spassky N, Pierani A, Causeret F. Repurposing of the multiciliation gene regulatory network in fate specification of Cajal-Retzius neurons. Dev Cell 2023; 58:1365-1382.e6. [PMID: 37321213 DOI: 10.1016/j.devcel.2023.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/06/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023]
Abstract
Cajal-Retzius cells (CRs) are key players in cerebral cortex development, and they display a unique transcriptomic identity. Here, we use scRNA-seq to reconstruct the differentiation trajectory of mouse hem-derived CRs, and we unravel the transient expression of a complete gene module previously known to control multiciliogenesis. However, CRs do not undergo centriole amplification or multiciliation. Upon deletion of Gmnc, the master regulator of multiciliogenesis, CRs are initially produced but fail to reach their normal identity resulting in their massive apoptosis. We further dissect the contribution of multiciliation effector genes and identify Trp73 as a key determinant. Finally, we use in utero electroporation to demonstrate that the intrinsic competence of hem progenitors as well as the heterochronic expression of Gmnc prevent centriole amplification in the CR lineage. Our work exemplifies how the co-option of a complete gene module, repurposed to control a distinct process, may contribute to the emergence of novel cell identities.
Collapse
Affiliation(s)
- Matthieu X Moreau
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Yoann Saillour
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Vicente Elorriaga
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Benoît Bouloudi
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Elodie Delberghe
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Tanya Deutsch Guerrero
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Amaia Ochandorena-Saa
- Université Paris Cité, Imagine-Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, 75015 Paris, France
| | - Laura Maeso-Alonso
- Instituto de Biomedicina, y Departamento de Biología Molecular, Universidad de León, 24071 Leon, Spain
| | - Margarita M Marques
- Instituto de Desarrollo Ganadero y Sanidad Animal, y Departamento de Producción Animal, Universidad de León, 24071 Leon, Spain
| | - Maria C Marin
- Instituto de Biomedicina, y Departamento de Biología Molecular, Universidad de León, 24071 Leon, Spain
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Alessandra Pierani
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Frédéric Causeret
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France.
| |
Collapse
|
3
|
Han JS, Fishman-Williams E, Decker SC, Hino K, Reyes RV, Brown NL, Simó S, Torre AL. Notch directs telencephalic development and controls neocortical neuron fate determination by regulating microRNA levels. Development 2023; 150:dev201408. [PMID: 37272771 PMCID: PMC10309580 DOI: 10.1242/dev.201408] [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: 10/27/2022] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
The central nervous system contains a myriad of different cell types produced from multipotent neural progenitors. Neural progenitors acquire distinct cell identities depending on their spatial position, but they are also influenced by temporal cues to give rise to different cell populations over time. For instance, the progenitors of the cerebral neocortex generate different populations of excitatory projection neurons following a well-known sequence. The Notch signaling pathway plays crucial roles during this process, but the molecular mechanisms by which Notch impacts progenitor fate decisions have not been fully resolved. Here, we show that Notch signaling is essential for neocortical and hippocampal morphogenesis, and for the development of the corpus callosum and choroid plexus. Our data also indicate that, in the neocortex, Notch controls projection neuron fate determination through the regulation of two microRNA clusters that include let-7, miR-99a/100 and miR-125b. Our findings collectively suggest that balanced Notch signaling is crucial for telencephalic development and that the interplay between Notch and miRNAs is essential for the control of neocortical progenitor behaviors and neuron cell fate decisions.
Collapse
Affiliation(s)
- Jisoo S. Han
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | | | - Steven C. Decker
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Keiko Hino
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Raenier V. Reyes
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Nadean L. Brown
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Sergi Simó
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
4
|
Nguyen H, Sokpor G, Parichha A, Pham L, Saikhedkar N, Xie Y, Ulmke PA, Rosenbusch J, Pirouz M, Behr R, Stoykova A, Brand-Saberi B, Nguyen HP, Staiger JF, Tole S, Tuoc T. BAF (mSWI/SNF) complex regulates mediolateral cortical patterning in the developing forebrain. Front Cell Dev Biol 2022; 10:1011109. [PMID: 36263009 PMCID: PMC9573979 DOI: 10.3389/fcell.2022.1011109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Early forebrain patterning entails the correct regional designation of the neuroepithelium, and appropriate specification, generation, and distribution of neural cells during brain development. Specific signaling and transcription factors are known to tightly regulate patterning of the dorsal telencephalon to afford proper structural/functional cortical arealization and morphogenesis. Nevertheless, whether and how changes of the chromatin structure link to the transcriptional program(s) that control cortical patterning remains elusive. Here, we report that the BAF chromatin remodeling complex regulates the spatiotemporal patterning of the mouse dorsal telencephalon. To determine whether and how the BAF complex regulates cortical patterning, we conditionally deleted the BAF complex scaffolding subunits BAF155 and BAF170 in the mouse dorsal telencephalic neuroepithelium. Morphological and cellular changes in the BAF mutant forebrain were examined using immunohistochemistry and in situ hybridization. RNA sequencing, Co-immunoprecipitation, and mass spectrometry were used to investigate the molecular basis of BAF complex involvement in forebrain patterning. We found that conditional ablation of BAF complex in the dorsal telencephalon neuroepithelium caused expansion of the cortical hem and medial cortex beyond their developmental boundaries. Consequently, the hippocampal primordium is not specified, the mediolateral cortical patterning is compromised, and the cortical identity is disturbed in the absence of BAF complex. The BAF complex was found to interact with the cortical hem suppressor LHX2. The BAF complex suppresses cortical hem fate to permit proper forebrain patterning. We provide evidence that BAF complex modulates mediolateral cortical patterning possibly by interacting with the transcription factor LHX2 to drive the LHX2-dependent transcriptional program essential for dorsal telencephalon patterning. Our data suggest a putative mechanistic synergy between BAF chromatin remodeling complex and LHX2 in regulating forebrain patterning and ontogeny.
Collapse
Affiliation(s)
- Huong Nguyen
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Faculty of Biotechnology, Thai Nguyen University of Sciences, Thai Nguyen, Vietnam
| | - Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | | | - Linh Pham
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | | | - Yuanbin Xie
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Pauline Antonie Ulmke
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Mehdi Pirouz
- Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, United States
| | - Rüdiger Behr
- German Primate Center-Leibniz Institute for Primate Research, Goettingen, Germany
| | | | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Jochen F. Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Shubha Tole
- Tata Institute of Fundamental Research, Mumbai, India
- *Correspondence: Shubha Tole, ; Tran Tuoc,
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Shubha Tole, ; Tran Tuoc,
| |
Collapse
|
5
|
Mercurio S, Serra L, Pagin M, Nicolis SK. Deconstructing Sox2 Function in Brain Development and Disease. Cells 2022; 11:cells11101604. [PMID: 35626641 PMCID: PMC9139651 DOI: 10.3390/cells11101604] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 02/04/2023] Open
Abstract
SOX2 is a transcription factor conserved throughout vertebrate evolution, whose expression marks the central nervous system from the earliest developmental stages. In humans, SOX2 mutation leads to a spectrum of CNS defects, including vision and hippocampus impairments, intellectual disability, and motor control problems. Here, we review how conditional Sox2 knockout (cKO) in mouse with different Cre recombinases leads to very diverse phenotypes in different regions of the developing and postnatal brain. Surprisingly, despite the widespread expression of Sox2 in neural stem/progenitor cells of the developing neural tube, some regions (hippocampus, ventral forebrain) appear much more vulnerable than others to Sox2 deletion. Furthermore, the stage of Sox2 deletion is also a critical determinant of the resulting defects, pointing to a stage-specificity of SOX2 function. Finally, cKOs illuminate the importance of SOX2 function in different cell types according to the different affected brain regions (neural precursors, GABAergic interneurons, glutamatergic projection neurons, Bergmann glia). We also review human genetics data regarding the brain defects identified in patients carrying mutations within human SOX2 and examine the parallels with mouse mutants. Functional genomics approaches have started to identify SOX2 molecular targets, and their relevance for SOX2 function in brain development and disease will be discussed.
Collapse
|
6
|
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
|
7
|
Nasu M, Esumi S, Hatakeyama J, Tamamaki N, Shimamura K. Two-Phase Lineage Specification of Telencephalon Progenitors Generated From Mouse Embryonic Stem Cells. Front Cell Dev Biol 2021; 9:632381. [PMID: 33937233 PMCID: PMC8086603 DOI: 10.3389/fcell.2021.632381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/09/2021] [Indexed: 12/16/2022] Open
Abstract
Proper brain development requires precisely controlled phases of stem cell proliferation, lineage specification, differentiation, and migration. Lineage specification depends partly on concentration gradients of chemical cues called morphogens. However, the rostral brain (telencephalon) expands prominently during embryonic development, dynamically altering local morphogen concentrations, and telencephalic subregional properties develop with a time lag. Here, we investigated how progenitor specification occurs under these spatiotemporally changing conditions using a three-dimensional in vitro differentiation model. We verified the critical contributions of three signaling factors for the lineage specification of subregional tissues in the telencephalon, ventralizing sonic hedgehog (Shh) and dorsalizing bone morphogenetic proteins (BMPs) and WNT proteins (WNTs). We observed that a short-lasting signal is sufficient to induce subregional progenitors and that the timing of signal exposure for efficient induction is specific to each lineage. Furthermore, early and late progenitors possess different Shh signal response capacities. This study reveals a novel developmental mechanism for telencephalon patterning that relies on the interplay of dose- and time-dependent signaling, including a time lag for specification and a temporal shift in cellular Shh sensitivity. This delayed fate choice through two-phase specification allows tissues with marked size expansion, such as the telencephalon, to compensate for the changing dynamics of morphogen signals.
Collapse
Affiliation(s)
- Makoto Nasu
- Department of Health Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeyuki Esumi
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Hatakeyama
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Nobuaki Tamamaki
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
8
|
Esteve P, Crespo I, Kaimakis P, Sandonís A, Bovolenta P. Sfrp1 Modulates Cell-signaling Events Underlying Telencephalic Patterning, Growth and Differentiation. Cereb Cortex 2020; 29:1059-1074. [PMID: 30084950 DOI: 10.1093/cercor/bhy013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 01/09/2018] [Indexed: 12/19/2022] Open
Abstract
The mammalian dorsal telencephalic neuroepithelium develops-from medial to lateral-into the choroid plaque, cortical hem, hippocampal primordium and isocortex under the influence of Bmp, Wnt and Notch signaling. Correct telencephalic development requires a tight coordination of the extent/duration of these signals, but the identification of possible molecular coordinators is still limited. Here, we postulated that Secreted Frizzled Related Protein 1 (Sfrp1), a multifunctional regulator of Bmp, Wnt and Notch signaling strongly expressed during early telencephalic development, may represent 1 of such molecules. We report that in E10.5-E12.5 Sfrp1-/- embryos, the hem and hippocampal domains are reduced in size whereas the prospective neocortex is medially extended. These changes are associated with a significant reduction of the medio-lateral telencephalic expression of Axin2, a read-out of Wnt/βcatenin signaling activation. Furthermore, in the absence of Sfrp1, Notch signaling is increased, cortical progenitor cell cycle is shorter, with expanded progenitor pools and enhanced generation of early-born neurons. Hence, in postnatal Sfrp1-/- animals the anterior hippocampus is reduced and the neocortex is shorter in the antero-posterior and medio-lateral axis but is thicker. We propose that, by controlling Wnt and Notch signaling in opposite directions, Sfrp1 promotes hippocampal patterning and balances medio-lateral and antero-posterior cortex expansion.
Collapse
Affiliation(s)
- Pilar Esteve
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Inmaculada Crespo
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Polynikis Kaimakis
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Africa Sandonís
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular "Severo Ochoa", CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), c/Nicolás Cabrera, Madrid, Spain
| |
Collapse
|
9
|
Sindhu SK, Udaykumar N, Zaidi MAA, Soni A, Sen J. MicroRNA-19b restricts Wnt7b to the hem, which induces aspects of hippocampus development in the avian forebrain. Development 2019; 146:146/20/dev175729. [PMID: 31578189 DOI: 10.1242/dev.175729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 08/27/2019] [Indexed: 01/21/2023]
Abstract
The functions of the hippocampus are conserved between birds and mammals; however, it is not known whether similar mechanisms are responsible for its development in these two classes. In mammals, hippocampus development is known to be regulated by the hem organizer. Here, we have identified that, in birds, Wnt7b secreted from the hem is sufficient for inducing the expression of hippocampal markers. Furthermore, we have demonstrated that a microRNA, miR-19b, which is selectively excluded from the hem region, is necessary and sufficient for restricting the expression of Wnt7b to the hem. This study suggests that the role of the Wnt signal emanating from the hem is conserved between birds and mammals, and that a microRNA-based mechanism is crucial for determining the position of the hippocampus.
Collapse
Affiliation(s)
- Suvimal Kumar Sindhu
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Niveda Udaykumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Mohd Ali Abbas Zaidi
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Abhinav Soni
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Jonaki Sen
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
10
|
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
|
11
|
Ruiz-Reig N, Andres B, Lamonerie T, Theil T, Fairén A, Studer M. The caudo-ventral pallium is a novel pallial domain expressing Gdf10 and generating Ebf3-positive neurons of the medial amygdala. Brain Struct Funct 2018; 223:3279-3295. [PMID: 29869132 DOI: 10.1007/s00429-018-1687-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/18/2018] [Indexed: 12/16/2022]
Abstract
In rodents, the medial nucleus of the amygdala receives direct inputs from the accessory olfactory bulbs and is mainly implicated in pheromone-mediated reproductive and defensive behaviors. The principal neurons of the medial amygdala are GABAergic neurons generated principally in the caudo-ventral medial ganglionic eminence and preoptic area. Beside GABAergic neurons, the medial amygdala also contains glutamatergic Otp-expressing neurons cells generated in the lateral hypothalamic neuroepithelium and a non-well characterized Pax6-positive population. In the present work, we describe a novel glutamatergic Ebf3-expressing neuronal subpopulation distributed within the periphery of the postero-ventral medial amygdala. These neurons are generated in a pallial domain characterized by high expression of Gdf10. This territory is topologically the most caudal tier of the ventral pallium and accordingly, we named it Caudo-Ventral Pallium (CVP). In the absence of Pax6, the CVP is disrupted and Ebf3-expressing neurons fail to be generated. Overall, this work proposes a novel model of the neuronal composition of the medial amygdala and unravels for the first time a new novel pallial subpopulation originating from the CVP and expressing the transcription factor Ebf3.
Collapse
Affiliation(s)
- Nuria Ruiz-Reig
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain.
| | - Belen Andres
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
| | - Thomas Lamonerie
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France
| | - Thomas Theil
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Alfonso Fairén
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
- , Palau 11, 03550, San Juan de Alicante, Spain
| | - Michèle Studer
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
| |
Collapse
|
12
|
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
|
13
|
Abstract
The formation of the nervous system is a multistep process that yields a mature brain. Failure in any of the steps of this process may cause brain malfunction. In the early stages of embryonic development, neural progenitors quickly proliferate and then, at a specific moment, differentiate into neurons or glia. Once they become postmitotic neurons, they migrate to their final destinations and begin to extend their axons to connect with other neurons, sometimes located in quite distant regions, to establish different neural circuits. During the last decade, it has become evident that Zic genes, in addition to playing important roles in early development (e.g., gastrulation and neural tube closure), are involved in different processes of late brain development, such as neuronal migration, axon guidance, and refinement of axon terminals. ZIC proteins are therefore essential for the proper wiring and connectivity of the brain. In this chapter, we review our current knowledge of the role of Zic genes in the late stages of neural circuit formation.
Collapse
|
14
|
Di Lullo E, Kriegstein AR. The use of brain organoids to investigate neural development and disease. Nat Rev Neurosci 2017; 18:573-584. [PMID: 28878372 PMCID: PMC5667942 DOI: 10.1038/nrn.2017.107] [Citation(s) in RCA: 521] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding the development and dysfunction of the human brain is a major goal of neurobiology. Much of our current understanding of human brain development has been derived from the examination of post-mortem and pathological specimens, bolstered by observations of developing non-human primates and experimental studies focused largely on mouse models. However, these tissue specimens and model systems cannot fully capture the unique and dynamic features of human brain development. Recent advances in stem cell technologies that enable the generation of human brain organoids from pluripotent stem cells (PSCs) promise to profoundly change our understanding of the development of the human brain and enable a detailed study of the pathogenesis of inherited and acquired brain diseases.
Collapse
Affiliation(s)
- Elizabeth Di Lullo
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California 94143, USA
- Department of Neurology, University of California, San Francisco, California 94158, USA
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California 94143, USA
- Department of Neurology, University of California, San Francisco, California 94158, USA
| |
Collapse
|
15
|
Mulligan KA, Cheyette BNR. Neurodevelopmental Perspectives on Wnt Signaling in Psychiatry. MOLECULAR NEUROPSYCHIATRY 2017; 2:219-246. [PMID: 28277568 DOI: 10.1159/000453266] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mounting evidence indicates that Wnt signaling is relevant to pathophysiology of diverse mental illnesses including schizophrenia, bipolar disorder, and autism spectrum disorder. In the 35 years since Wnt ligands were first described, animal studies have richly explored how downstream Wnt signaling pathways affect an array of neurodevelopmental processes and how their disruption can lead to both neurological and behavioral phenotypes. Recently, human induced pluripotent stem cell (hiPSC) models have begun to contribute to this literature while pushing it in increasingly translational directions. Simultaneously, large-scale human genomic studies are providing evidence that sequence variation in Wnt signal pathway genes contributes to pathogenesis in several psychiatric disorders. This article reviews neurodevelopmental and postneurodevelopmental functions of Wnt signaling, highlighting mechanisms, whereby its disruption might contribute to psychiatric illness, and then reviews the most reliable recent genetic evidence supporting that mutations in Wnt pathway genes contribute to psychiatric illness. We are proponents of the notion that studies in animal and hiPSC models informed by the human genetic data combined with the deep knowledge base and tool kits generated over the last several decades of basic neurodevelopmental research will yield near-term tangible advances in neuropsychiatry.
Collapse
Affiliation(s)
- Kimberly A Mulligan
- Department of Biological Sciences, California State University, Sacramento, CA, USA
| | - Benjamin N R Cheyette
- Department of Psychiatry, Kavli Institute for Fundamental Neuroscience, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
16
|
Tuoc T, Dere E, Radyushkin K, Pham L, Nguyen H, Tonchev AB, Sun G, Ronnenberg A, Shi Y, Staiger JF, Ehrenreich H, Stoykova A. Ablation of BAF170 in Developing and Postnatal Dentate Gyrus Affects Neural Stem Cell Proliferation, Differentiation, and Learning. Mol Neurobiol 2016; 54:4618-4635. [PMID: 27392482 PMCID: PMC5509785 DOI: 10.1007/s12035-016-9948-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 06/03/2016] [Indexed: 10/27/2022]
Abstract
The BAF chromatin remodeling complex plays an essential role in brain development. However its function in postnatal neurogenesis in hippocampus is still unknown. Here, we show that in postnatal dentate gyrus (DG), the BAF170 subunit of the complex is expressed in radial glial-like (RGL) progenitors and in cell types involved in subsequent steps of adult neurogenesis including mature astrocytes. Conditional deletion of BAF170 during cortical late neurogenesis as well as during adult brain neurogenesis depletes the pool of RGL cells in DG, and promotes terminal astrocyte differentiation. These derangements are accompanied by distinct behavioral deficits, as reflected by an impaired accuracy of place responding in the Morris water maze test, during both hidden platform as well as reversal learning. Inducible deletion of BAF170 in DG during adult brain neurogenesis resulted in mild spatial learning deficits, having a more pronounced effect on spatial learning during the reversal test. These findings demonstrate involvement of BAF170-dependent chromatin remodeling in hippocampal neurogenesis and cognition and suggest a specific role of adult neurogenesis in DG in adaptive behavior.
Collapse
Affiliation(s)
- Tran Tuoc
- Institute of Neuroanatomy, University Medical Center, Georg-August University Göttingen, Göttingen, Germany. .,Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany. .,DFG Center for Nanoscale Microscopy & Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.
| | - Ekrem Dere
- DFG Center for Nanoscale Microscopy & Molecular Physiology of the Brain (CNMPB), Göttingen, Germany. .,Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| | - Konstantin Radyushkin
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Linh Pham
- Institute of Neuroanatomy, University Medical Center, Georg-August University Göttingen, Göttingen, Germany
| | - Huong Nguyen
- Institute of Neuroanatomy, University Medical Center, Georg-August University Göttingen, Göttingen, Germany
| | - Anton B Tonchev
- Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany.,Department of Anatomy, Histology and Embryology, Medical University of Varna, Varna, Bulgaria
| | - Guoqiang Sun
- Division of Stem Cell Biology Research, Department of Developmental and Stem Cell Biology, Cancer Center, Beckman Research Institute of City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Anja Ronnenberg
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Yanhong Shi
- Division of Stem Cell Biology Research, Department of Developmental and Stem Cell Biology, Cancer Center, Beckman Research Institute of City of Hope, 1500 E. Duarte Road, Duarte, CA, 91010, USA
| | - Jochen F Staiger
- Institute of Neuroanatomy, University Medical Center, Georg-August University Göttingen, Göttingen, Germany.,DFG Center for Nanoscale Microscopy & Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Hannelore Ehrenreich
- DFG Center for Nanoscale Microscopy & Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.,Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Anastassia Stoykova
- Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany. .,DFG Center for Nanoscale Microscopy & Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.
| |
Collapse
|
17
|
Ypsilanti AR, Rubenstein JLR. Transcriptional and epigenetic mechanisms of early cortical development: An examination of how Pax6 coordinates cortical development. J Comp Neurol 2015; 524:609-29. [PMID: 26304102 DOI: 10.1002/cne.23866] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 07/14/2015] [Accepted: 07/17/2015] [Indexed: 12/26/2022]
Abstract
The development of the cortex is an elaborate process that integrates a plethora of finely tuned molecular processes ranging from carefully regulated gradients of transcription factors, dynamic changes in the chromatin landscape, or formation of protein complexes to elicit and regulate transcription. Combined with cellular processes such as cell type specification, proliferation, differentiation, and migration, all of these developmental processes result in the establishment of an adult mammalian cortex with its typical lamination and regional patterning. By examining in-depth the role of one transcription factor, Pax6, on the regulation of cortical development, its integration in the regulation of chromatin state, and its regulation by cis-regulatory elements, we aim to demonstrate the importance of integrating each level of regulation in our understanding of cortical development.
Collapse
Affiliation(s)
- Athéna R Ypsilanti
- Department of Psychiatry, Neuroscience Program, and the Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, California
| | - John L R Rubenstein
- Department of Psychiatry, Neuroscience Program, and the Nina Ireland Laboratory of Developmental Neurobiology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
18
|
Hevner RF. Evolution of the mammalian dentate gyrus. J Comp Neurol 2015; 524:578-94. [PMID: 26179319 DOI: 10.1002/cne.23851] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/02/2015] [Accepted: 07/06/2015] [Indexed: 01/08/2023]
Abstract
The dentate gyrus (DG), a part of the hippocampal formation, has important functions in learning, memory, and adult neurogenesis. Compared with homologous areas in sauropsids (birds and reptiles), the mammalian DG is larger and exhibits qualitatively different phenotypes: 1) folded (C- or V-shaped) granule neuron layer, concave toward the hilus and delimited by a hippocampal fissure; 2) nonperiventricular adult neurogenesis; and 3) prolonged ontogeny, involving extensive abventricular (basal) migration and proliferation of neural stem and progenitor cells (NSPCs). Although gaps remain, available data indicate that these DG traits are present in all orders of mammals, including monotremes and marsupials. The exception is Cetacea (whales, dolphins, and porpoises), in which DG size, convolution, and adult neurogenesis have undergone evolutionary regression. Parsimony suggests that increased growth and convolution of the DG arose in stem mammals concurrently with nonperiventricular adult hippocampal neurogenesis and basal migration of NSPCs during development. These traits could all result from an evolutionary change that enhanced radial migration of NSPCs out of the periventricular zones, possibly by epithelial-mesenchymal transition, to colonize and maintain nonperiventricular proliferative niches. In turn, increased NSPC migration and clonal expansion might be a consequence of growth in the cortical hem (medial patterning center), which produces morphogens such as Wnt3a, generates Cajal-Retzius neurons, and is regulated by Lhx2. Finally, correlations between DG convolution and neocortical gyrification (or capacity for gyrification) suggest that enhanced abventricular migration and proliferation of NSPCs played a transformative role in growth and folding of neocortex as well as archicortex.
Collapse
Affiliation(s)
- Robert F Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington, 98101
- Department of Neurological Surgery, University of Washington, Seattle, Washington, 98104
| |
Collapse
|
19
|
Seki T, Sato T, Toda K, Osumi N, Imura T, Shioda S. Distinctive population of Gfap-expressing neural progenitors arising around the dentate notch migrate and form the granule cell layer in the developing hippocampus. J Comp Neurol 2014; 522:261-83. [PMID: 23983092 DOI: 10.1002/cne.23460] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/12/2013] [Accepted: 08/14/2013] [Indexed: 01/24/2023]
Abstract
In the adult hippocampus, granule cells continue to be generated from astrocyte-like progenitors expressing glial fibrillary acidic protein (GFAP) that differ from embryonic neocortical progenitors. However, during the embryonic period, dentate granule neurons and neocortical pyramidal neurons are derived from the ventricular zone (VZ) of the pallium. Our question is when do GFAP+ progenitors of granule neurons appear in the developing hippocampus during the embryonic period, and how do they form the granule cell layer. The present analysis using Gfap-GFP transgenic mice shows that the GFP+ distinct cell population first appears in the VZ of the medial pallium at the dorsal edge of the fimbria on embryonic day 13.5. During the perinatal period, they form a migratory stream from the VZ to the developing dentate gyrus, and establish the germinal zones in the migratory stream, and the marginal and hilar regions in the developing dentate gyrus. GFP+ cells in these regions were positive for Sox2 and Ki67, but negative for BLBP. GFP+ cells with Neurogenin2 expression were largely distributed in the VZ, whereas GFP+ cells with Tbr2 and NeuroD expressions were seen in the migratory stream and developing dentate gyrus. Prox1-expressing GFP+ cells were restricted to the developing dentate gyrus. These results suggest that distinctive Gfap-expressing progenitors arising around the dentate notch form germinal regions in the migratory stream and the developing dentate gyrus where they differentiate into granule neurons, indicating that distinct astrocyte-like neural progenitors continue to generate granule neurons, from the beginning of dentate development and throughout life. J. Comp. Neurol. 522:261-283, 2014. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, 160-8402, Japan
| | | | | | | | | | | |
Collapse
|
20
|
Belvindrah R, Nosten-Bertrand M, Francis F. Neuronal migration and its disorders affecting the CA3 region. Front Cell Neurosci 2014; 8:63. [PMID: 24624057 PMCID: PMC3941003 DOI: 10.3389/fncel.2014.00063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 02/13/2014] [Indexed: 11/15/2022] Open
Abstract
In this review, we focus on CA3 neuronal migration disorders in the rodent. We begin by introducing the main steps of hippocampal development, and we summarize characteristic hippocampal malformations in human. We then describe various mouse mutants showing structural hippocampal defects. Notably, genes identified in human cortical neuronal migration disorders consistently give rise to a CA3 phenotype when mutated in the mouse. We successively describe their molecular, physiological and behavioral phenotypes that together contribute to a better understanding of CA3-dependent functions. We finally discuss potential factors underlying the CA3 vulnerability revealed by these mouse mutants and that may also contribute to other human neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Richard Belvindrah
- INSERM UMR-S 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06 Paris, France ; Institut du Fer à Moulin Paris, France
| | - Marika Nosten-Bertrand
- INSERM UMR-S 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06 Paris, France ; Institut du Fer à Moulin Paris, France
| | - Fiona Francis
- INSERM UMR-S 839 Paris, France ; Sorbonne Universités, Université Pierre et Marie Curie, Univ Paris 06 Paris, France ; Institut du Fer à Moulin Paris, France
| |
Collapse
|
21
|
Li G, Pleasure SJ. The development of hippocampal cellular assemblies. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 3:165-77. [PMID: 24719288 DOI: 10.1002/wdev.127] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Revised: 08/19/2013] [Accepted: 08/29/2013] [Indexed: 11/10/2022]
Abstract
The proper assembly of a cohort of distinct cell types is a prerequisite for building a functional hippocampus. In this review, we describe the major molecular events of the developmental program leading to the cellular construction of the hippocampus. Data from rodent studies are used here to elaborate on our understanding of these processes.
Collapse
Affiliation(s)
- Guangnan Li
- Department of Neurology, Programs in Neuroscience and Developmental Biology, Institute for Regenerative Medicine, University of California, San Francisco, CA, USA
| | | |
Collapse
|
22
|
Stephen LA, Davis GM, McTeir KE, James J, McTeir L, Kierans M, Bain A, Davey MG. Failure of centrosome migration causes a loss of motile cilia in talpid(3) mutants. Dev Dyn 2013; 242:923-31. [PMID: 23613203 DOI: 10.1002/dvdy.23980] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/26/2013] [Accepted: 04/13/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Loss of function mutations in the centrosomal protein TALPID3 (KIAA0586) cause a failure of primary cilia formation in animal models and are associated with defective Hedgehog signalling. It is unclear, however, if TALPID3 is required only for primary cilia formation or if it is essential for all ciliogenesis, including that of motile cilia in multiciliate cells. RESULTS FOXJ1, a key regulator of multiciliate cell fate, is expressed in the dorsal neuroectoderm of the chicken forebrain and hindbrain at stage 20HH, in areas that will give rise to choroid plexuses in both wt and talpid(3) embryos. Wt ependymal cells of the prosencephalic choroid plexuses subsequently transition from exhibiting single short cilia to multiple long motile cilia at 29HH (E8). Primary cilia and long motile cilia were only rarely observed on talpid(3) ependymal cells. Electron microscopy determined that talpid(3) ependymal cells do develop multiple centrosomes in accordance with FOXJ1 expression, but these fail to migrate to the apical surface of ependymal cells although axoneme formation was sometimes observed. CONCLUSIONS TALPID3, which normally localises to the proximal centrosome, is essential for centrosomal migration prior to ciliogenesis but is not directly required for de novo centriologenesis, multiciliated fate, or axoneme formation.
Collapse
Affiliation(s)
- Louise A Stephen
- Division of Developmental Biology, The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Khalaf-Nazzal R, Francis F. Hippocampal development - old and new findings. Neuroscience 2013; 248:225-42. [PMID: 23756184 DOI: 10.1016/j.neuroscience.2013.05.061] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 05/14/2013] [Accepted: 05/31/2013] [Indexed: 01/26/2023]
Abstract
The hippocampus, derived from medial regions of the telencephalon, constitutes a remarkable brain structure. It is part of the limbic system, and it plays important roles in information encoding, related to short-term and long-term memory, and spatial navigation. It has also attracted the attention of many clinicians and neuroscientists for its involvement in a wide spectrum of pathological conditions, including epilepsy, intellectual disability, Alzheimer disease and others. Here we address the topic of hippocampal development. As well as original landmark findings, modern techniques such as large-scale in situ hybridizations, in utero electroporation and the study of mouse mutants with hippocampal phenotypes, add further detail to our knowledge of the finely regulated processes which form this intricate structure. Molecular signatures are being revealed related to field, intra-field and laminar cell identity, as well as, cell compartments expressing surface proteins instrumental for connectivity. We summarize here old and new findings, and highlight elegant tools used to fine-study hippocampal development.
Collapse
Affiliation(s)
- R Khalaf-Nazzal
- INSERM, UMR-S 839, Paris 75005, France; Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France
| | - F Francis
- INSERM, UMR-S 839, Paris 75005, France; Université Pierre et Marie Curie, Paris 75005, France; Institut du Fer à Moulin, Paris 75005, France.
| |
Collapse
|
24
|
Alfano C, Studer M. Neocortical arealization: evolution, mechanisms, and open questions. Dev Neurobiol 2013; 73:411-47. [PMID: 23239642 DOI: 10.1002/dneu.22067] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 11/03/2012] [Accepted: 12/06/2012] [Indexed: 12/13/2022]
Abstract
The mammalian neocortex is a structure with no equals in the vertebrates and is the seat of the highest cerebral functions, such as thoughts and consciousness. It is radially organized into six layers and tangentially subdivided into functional areas deputed to the elaboration of sensory information, association between different stimuli, and selection and triggering of voluntary movements. The process subdividing the neocortical field into several functional areas is called "arealization". Each area has its own cytoarchitecture, connectivity, and peculiar functions. In the last century, several neuroscientists have investigated areal structure and the mechanisms that have led during evolution to the rising of the neocortex and its organization. The extreme conservation in the positioning and wiring of neocortical areas among different mammalian families suggests a conserved genetic program orchestrating neocortical patterning. However, the impressive plasticity of the neocortex, which is able to rewire and reorganize areal structures and connectivity after impairments of sensory pathways, argues for a more complex scenario. Indeed, even if genetics and molecular biology helped in identifying several genes involved in the arealization process, the logic underlying the neocortical bauplan is still beyond our comprehension. In this review, we will introduce the present knowledge and hypotheses on the ontogenesis and evolution of neocortical areas. Then, we will focus our attention on some open issues, which are still unresolved, and discuss some recent studies that might open new directions to be explored in the next few years.
Collapse
Affiliation(s)
- Christian Alfano
- Institute of Biology Valrose, iBV, UMR INSERM1091/CNRS7277/UNS, Nice, F-06108, France.
| | | |
Collapse
|
25
|
Abstract
Atypical cadherin Celsr3, a regulator of planar cell polarity, is critical for the development of the axonal blueprint. We previously showed that expression of Celsr3 is necessary to establish forebrain connections such as the anterior commissure and thalamocortical and corticospinal tracts. The requirement for Celsr3 during hippocampal wiring and its action in the hippocampus remain largely unexplored. Here, we compared the connectivity and maturation of the hippocampal formation in Celsr3|Foxg1 and Celsr3|Dlx mice. Celsr3 is inactivated in the whole telencephalon, including the hippocampal primordium, in Celsr3|Foxg1 mice, and in the early basal telencephalon, including ganglionic eminences and ventral diencephalon, in Celsr3|Dlx mice. Behavioral tests showed that both mutants were hyperactive and had impaired learning and memory. Abnormal cytoarchitecture of CA1, CA3, and dentate gyrus was found in the Celsr3|Foxg1 mutant, in which afferent and efferent hippocampal pathways, as well as intrinsic connections, were dramatically disrupted. In Celsr3|Dlx mutant mice, hippocampal cytoarchitecture was mildly affected and extrinsic and intrinsic connectivity moderately disturbed. In both mutants, pyramidal neurons in CA1 harbored atrophic dendritic trees, with decreased synapse density and increased proportion of symmetric versus asymmetric synapses, and long-term potentiation was altered. In contrast, mutant hippocampal neurons extended neurites that were normal, even longer than those of control neurons, indicating that anomalies in vivo are secondary to defective connections. Postnatal neurogenesis was preserved and mutant interneurons were able to migrate to the hippocampus. Thus, like in neocortex, Celsr3 is required for hippocampal development, connectivity and function, and for pyramidal cell maturation.
Collapse
|
26
|
Suzuki IK, Hirata T. Neocortical neurogenesis is not really “neo”: A new evolutionary model derived from a comparative study of chick pallial development. Dev Growth Differ 2012; 55:173-87. [DOI: 10.1111/dgd.12020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 10/18/2012] [Accepted: 10/18/2012] [Indexed: 12/24/2022]
Affiliation(s)
- Ikuo K. Suzuki
- Division of Brain Function; National Institute of Genetics; Graduate University for Advanced Studies (Sokendai); Yata 1111; Mishima; 411-8540; Japan
| | - Tatsumi Hirata
- Division of Brain Function; National Institute of Genetics; Graduate University for Advanced Studies (Sokendai); Yata 1111; Mishima; 411-8540; Japan
| |
Collapse
|
27
|
Konno D, Iwashita M, Satoh Y, Momiyama A, Abe T, Kiyonari H, Matsuzaki F. The mammalian DM domain transcription factor Dmrta2 is required for early embryonic development of the cerebral cortex. PLoS One 2012; 7:e46577. [PMID: 23056351 PMCID: PMC3462758 DOI: 10.1371/journal.pone.0046577] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Accepted: 08/31/2012] [Indexed: 01/08/2023] Open
Abstract
Development of the mammalian telencephalon is precisely organized by a combination of extracellular signaling events derived from signaling centers and transcription factor networks. Using gene expression profiling of the developing mouse dorsal telencephalon, we found that the DM domain transcription factor Dmrta2 (doublesex and mab-3-related transcription factor a2) is involved in the development of the dorsal telencephalon. Consistent with its medial-high/lateral-low expression pattern in the dorsal telencephalon, Dmrta2 null mutants demonstrated a dramatic reduction in medial cortical structures such as the cortical hem and the choroid plexus, and a complete loss of the hippocampus. In this mutant, the dorsal telencephalon also showed a remarkable size reduction, in addition to abnormal cell cycle kinetics and defective patterning. In contrast, a conditional Dmrta2 deletion in the telencephalon, which was accomplished after entry into the neurogenic phase, resulted in only a slight reduction in telencephalon size and normal patterning. We also found that Dmrta2 expression was decreased by a dominant-negative Tcf and was increased by a stabilized β-catenin form. These data suggest that Dmrta2 plays pivotal roles in the early development of the telencephalon via the formation of the cortical hem, a source of Wnts, and also in the maintenance of neural progenitors as a downstream of the Wnt pathway.
Collapse
Affiliation(s)
- Daijiro Konno
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
- * E-mail: (DK); (FM)
| | - Misato Iwashita
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
| | - Yoshiaki Satoh
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
| | - Asuka Momiyama
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
- * E-mail: (DK); (FM)
| |
Collapse
|
28
|
Mulligan KA, Cheyette BNR. Wnt signaling in vertebrate neural development and function. J Neuroimmune Pharmacol 2012; 7:774-87. [PMID: 23015196 DOI: 10.1007/s11481-012-9404-x] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 09/10/2012] [Indexed: 02/03/2023]
Abstract
Members of the Wnt family of secreted signaling proteins influence many aspects of neural development and function. Wnts are required from neural induction and axis formation to axon guidance and synapse development, and even help modulate synapse activity. Wnt proteins activate a variety of downstream signaling pathways and can induce a similar variety of cellular responses, including gene transcription changes and cytoskeletal rearrangements. This review provides an introduction to Wnt signaling pathways and discusses current research on their roles in vertebrate neural development and function.
Collapse
Affiliation(s)
- Kimberly A Mulligan
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
29
|
Kheradmand Kia S, Verbeek E, Engelen E, Schot R, Poot R, de Coo I, Lequin M, Poulton C, Pourfarzad F, Grosveld F, Brehm A, de Wit M, Oegema R, Dobyns W, Verheijen F, Mancini G. RTTN mutations link primary cilia function to organization of the human cerebral cortex. Am J Hum Genet 2012; 91:533-40. [PMID: 22939636 DOI: 10.1016/j.ajhg.2012.07.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/09/2012] [Accepted: 07/11/2012] [Indexed: 10/28/2022] Open
Abstract
Polymicrogyria is a malformation of the developing cerebral cortex caused by abnormal organization and characterized by many small gyri and fusion of the outer molecular layer. We have identified autosomal-recessive mutations in RTTN, encoding Rotatin, in individuals with bilateral diffuse polymicrogyria from two separate families. Rotatin determines early embryonic axial rotation, as well as anteroposterior and dorsoventral patterning in the mouse. Human Rotatin has recently been identified as a centrosome-associated protein. The Drosophila melanogaster homolog of Rotatin, Ana3, is needed for structural integrity of centrioles and basal bodies and maintenance of sensory neurons. We show that Rotatin colocalizes with the basal bodies at the primary cilium. Cultured fibroblasts from affected individuals have structural abnormalities of the cilia and exhibit downregulation of BMP4, WNT5A, and WNT2B, which are key regulators of cortical patterning and are expressed at the cortical hem, the cortex-organizing center that gives rise to Cajal-Retzius (CR) neurons. Interestingly, we have shown that in mouse embryos, Rotatin colocalizes with CR neurons at the subpial marginal zone. Knockdown experiments in human fibroblasts and neural stem cells confirm a role for RTTN in cilia structure and function. RTTN mutations therefore link aberrant ciliary function to abnormal development and organization of the cortex in human individuals.
Collapse
|
30
|
Robertshaw E, Kiecker C. Phylogenetic origins of brain organisers. SCIENTIFICA 2012; 2012:475017. [PMID: 24278699 PMCID: PMC3820451 DOI: 10.6064/2012/475017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 06/21/2012] [Indexed: 06/02/2023]
Abstract
The regionalisation of the nervous system begins early in embryogenesis, concomitant with the establishment of the anteroposterior (AP) and dorsoventral (DV) body axes. The molecular mechanisms that drive axis induction appear to be conserved throughout the animal kingdom and may be phylogenetically older than the emergence of bilateral symmetry. As a result of this process, groups of patterning genes that are equally well conserved are expressed at specific AP and DV coordinates of the embryo. In the emerging nervous system of vertebrate embryos, this initial pattern is refined by local signalling centres, secondary organisers, that regulate patterning, proliferation, and axonal pathfinding in adjacent neuroepithelium. The main secondary organisers for the AP neuraxis are the midbrain-hindbrain boundary, zona limitans intrathalamica, and anterior neural ridge and for the DV neuraxis the notochord, floor plate, and roof plate. A search for homologous secondary organisers in nonvertebrate lineages has led to controversy over their phylogenetic origins. Based on a recent study in hemichordates, it has been suggested that the AP secondary organisers evolved at the base of the deuterostome superphylum, earlier than previously thought. According to this view, the lack of signalling centres in some deuterostome lineages is likely to reflect a secondary loss due to adaptive processes. We propose that the relative evolutionary flexibility of secondary organisers has contributed to a broader morphological complexity of nervous systems in different clades.
Collapse
Affiliation(s)
- Ellen Robertshaw
- MRC Centre for Developmental Neurobiology, King's College London, 4th Floor, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, UK
| | - Clemens Kiecker
- MRC Centre for Developmental Neurobiology, King's College London, 4th Floor, New Hunt's House, Guy's Hospital Campus, London SE1 1UL, UK
| |
Collapse
|
31
|
Rosenthal EH, Tonchev AB, Stoykova A, Chowdhury K. Regulation of archicortical arealization by the transcription factor Zbtb20. Hippocampus 2012; 22:2144-56. [PMID: 22689450 DOI: 10.1002/hipo.22035] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2012] [Indexed: 12/20/2022]
Abstract
The molecular mechanisms of regionalization of the medial pallium (MP), the anlage of the hippocampus, and transitional (cingulate and retrosplenial) cortices are largely unknown. Previous analyses have outlined an important role of the transcription factor (TF) Zbtb20 for hippocampal CA1 field specification (Nielsen et al. (2007) Development 134:1133-1140; Nielsen et al. (2010) Cereb Cortex 20:1904-1914; Xie et al. (2010) Proc Natl Acad Sci USA 107:6510-6515). Here, we present novel data showing that Zbtb20 exhibits a ventral(high)-to-dorsal(low) gradient of expression in MP progenitors as well as an expression in postmitotic cells at the transitional cortex/neocortex border. Our detailed pattern analysis revealed that in Zbtb20 loss-of-function the molecular borders between neocortical, transitional, and hippocampal fields are progressively shifted ventrally, leading to an ectopic positioning of all dorsal fields into the neighboring ventrally located areas. Thus, in addition to its known importance for the specification of the hippocampal CA1 sector, the graded expression of TF Zbtb20 in ventricular zone of MP appears to translate early positional information for establishment of all developing MP fields. Our data also suggest that the signaling factor Wnt3a is a putative molecular partner of TF Zbtb20 in this patterning process.
Collapse
Affiliation(s)
- Eva H Rosenthal
- Max Planck Institute for Biophysical Chemistry, Am Fassberg, Goettingen, Germany
| | | | | | | |
Collapse
|
32
|
Barkovich AJ, Guerrini R, Kuzniecky RI, Jackson GD, Dobyns WB. A developmental and genetic classification for malformations of cortical development: update 2012. Brain 2012; 135:1348-69. [PMID: 22427329 PMCID: PMC3338922 DOI: 10.1093/brain/aws019] [Citation(s) in RCA: 698] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Malformations of cerebral cortical development include a wide range of developmental disorders that are common causes of neurodevelopmental delay and epilepsy. In addition, study of these disorders contributes greatly to the understanding of normal brain development and its perturbations. The rapid recent evolution of molecular biology, genetics and imaging has resulted in an explosive increase in our knowledge of cerebral cortex development and in the number and types of malformations of cortical development that have been reported. These advances continue to modify our perception of these malformations. This review addresses recent changes in our perception of these disorders and proposes a modified classification based upon updates in our knowledge of cerebral cortical development.
Collapse
Affiliation(s)
- A James Barkovich
- Neuroradiology, University of California at San Francisco, 505 Parnassus Avenue, San Francisco, CA 94913-0628, USA.
| | | | | | | | | |
Collapse
|
33
|
Fotaki V, Price DJ, Mason JO. Wnt/β-catenin signaling is disrupted in the extra-toes (Gli3(Xt/Xt) ) mutant from early stages of forebrain development, concomitant with anterior neural plate patterning defects. J Comp Neurol 2012; 519:1640-57. [PMID: 21452227 DOI: 10.1002/cne.22592] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The zinc finger transcription factor Gli3 is essential for normal development of the forebrain. Mutant mice with no functional Gli3 (extra-toes, Gli3(Xt/Xt) mutants) display a massive reduction in the size of the telencephalic lobes and absence of dorsomedial telencephalic structures, including the cortical hem, which normally expresses a number of Wnt molecules essential for patterning the hippocampus. Dorsomedial telencephalic Wnt activity, transduced through the Wnt/β-catenin signaling pathway, is also required for hippocampal specification and dorsoventral telencephalic patterning. Wnts whose normal expression is restricted to the cortical hem are completely absent in Gli3(Xt/Xt) embryos, but some expression of those Wnts with a broader expression domain persists, raising the possibility that Wnt/β-catenin signaling may still be active in this mutant. We examined whether the Wnt expression that persists in the Gli3(Xt/Xt) mutant neocortex activates Wnt/β-catenin signaling, using the BAT-gal transgenic reporter. We found Wnt/β-catenin signaling consistently decreased in the forebrains of Gli3(Xt/Xt) mutants, even prior to the formation of the cortical hem. This is accompanied by a severe reduction in expression of Wnt7b and Wnt8b at the lateral edges of the anterior neural plate that will give rise to the pallium. In addition, we found a significant increase in the expression of rostroventral markers of the anterior neural plate that will give rise to the basal forebrain. Our data reveal that Gli3 is required at the neural plate stage to regulate Wnt expression and Wnt/β-catenin signaling in the presumptive forebrain and confirm its previously proposed role in patterning the anterior neural plate.
Collapse
Affiliation(s)
- Vassiliki Fotaki
- Genes and Development Group, Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9XD, United Kingdom.
| | | | | |
Collapse
|
34
|
|
35
|
Genetic and developmental homology in amniote brains. Toward conciliating radical views of brain evolution. Brain Res Bull 2010; 84:125-36. [PMID: 21146594 DOI: 10.1016/j.brainresbull.2010.12.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 10/26/2010] [Accepted: 12/05/2010] [Indexed: 11/23/2022]
Abstract
The six-layered neocortex is both a unique and a universal character of mammals. Historically, a major concern has been to determine its phylogenetic origins by establishing which structures, if any, correspond to it in the brains of other vertebrates. Two opposing hypotheses have been debated in the last years: (i) the neocortex arises entirely from the dorsal hemisphere of ancestral reptiles, and (ii) a large portion of it originates in the lateral hemisphere, from a structure termed the dorsal ventricular ridge (DVR), which expands significantly in reptiles and especially in birds. While developmental and genetic evidence strongly favors a dorsal origin of the neocortex, there are important similarities in the sensory connectivity to the neocortex and to the DVR, and more recently, in the phenotype of late-produced elements in both structures. It is proposed that, despite originating in different embryonic domains, the proliferative expansion of both the mammalian neocortex and the sauropsidian DVR is partly based on the amplification of similar developmental programs, possibly dependent on Pax6 activity or of related cascades that promote progenitor proliferation. While Pax6 activity is already present in the amphibian pallium, I propose that at some point(s) in amniote evolution it has been upregulated yielding brain expansion in both sauropsids and mammals. However, in the latter there has been an additional dorsalizing influence contributing to the development of the neocortex and restricting the expansion of the lateral hemisphere. Finally, a significant contribution to neocortical origins by anterior signaling centers secreting FGFs is suggested, by virtue of their association to olfactory development and their cortical patterning functions. This perspective fits a dynamical view of brain homology, where instead of searching for a one-to-one correspondence between components, emphasis is placed on changes in the modulation of conserved signaling centers and their corresponding morphogen gradients across species.
Collapse
|
36
|
Nielsen JV, Blom JB, Noraberg J, Jensen NA. Zbtb20-induced CA1 pyramidal neuron development and area enlargement in the cerebral midline cortex of mice. ACTA ACUST UNITED AC 2009; 20:1904-14. [PMID: 19955470 DOI: 10.1093/cercor/bhp261] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Expression of the transcriptional repressor Zbtb20 is confined to the hippocampal primordium of the developing dorsal midline cortex in mice. Here, we show that misexpression of Zbtb20 converts projection neurons of the subiculum and postsubiculum (dorsal presubiculum) to CA1 pyramidal neurons that are innervated by Schaffer collateral projections in ectopic strata oriens and radiatum. The Zbtb20-transformed neurons express Bcl11B, Satb2, and Calbindin-D28k, which are markers of adult CA1 pyramidal neurons. Downregulation of Zbtb20 expression by RNA interference impairs the normal maturation of CA1 pyramidal neurons resulting in deficiencies in Calbindin-D28k expression and in reduced apical dendritic arborizations in stratum lacunosum moleculare. Overall, the results show that Zbtb20 is required for various aspects of CA1 pyramidal neuron development such as the postnatal extension of apical dendritic arbors in the distal target zone and the subtype differentiation of Calbindin-D28k-positive subsets. They further suggest that Zbtb20 plays a role in arealization of the midline cortex.
Collapse
Affiliation(s)
- Jakob V Nielsen
- Molecular Neurobiology Laboratory, Medical Biotechnology Center, Department of Molecular Medicine, University of Southern Denmark, J.B. Winslows Vej 25, DK-5000 Odense C, Denmark
| | | | | | | |
Collapse
|