1
|
Brooks ER, Moorman AR, Bhattacharya B, Prudhomme IS, Land M, Alcorn HL, Sharma R, Pe'er D, Zallen JA. A single-cell atlas of spatial and temporal gene expression in the mouse cranial neural plate. eLife 2025; 13:RP102819. [PMID: 40192104 PMCID: PMC11975377 DOI: 10.7554/elife.102819] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
The formation of the mammalian brain requires regionalization and morphogenesis of the cranial neural plate, which transforms from an epithelial sheet into a closed tube that provides the structural foundation for neural patterning and circuit formation. Sonic hedgehog (SHH) signaling is important for cranial neural plate patterning and closure, but the transcriptional changes that give rise to the spatially regulated cell fates and behaviors that build the cranial neural tube have not been systematically analyzed. Here, we used single-cell RNA sequencing to generate an atlas of gene expression at six consecutive stages of cranial neural tube closure in the mouse embryo. Ordering transcriptional profiles relative to the major axes of gene expression predicted spatially regulated expression of 870 genes along the anterior-posterior and mediolateral axes of the cranial neural plate and reproduced known expression patterns with over 85% accuracy. Single-cell RNA sequencing of embryos with activated SHH signaling revealed distinct SHH-regulated transcriptional programs in the developing forebrain, midbrain, and hindbrain, suggesting a complex interplay between anterior-posterior and mediolateral patterning systems. These results define a spatiotemporally resolved map of gene expression during cranial neural tube closure and provide a resource for investigating the transcriptional events that drive early mammalian brain development.
Collapse
Affiliation(s)
- Eric R Brooks
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State UniversityRaleighUnited States
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Andrew R Moorman
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Bhaswati Bhattacharya
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Ian S Prudhomme
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Max Land
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Heather L Alcorn
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Roshan Sharma
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Dana Pe'er
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| |
Collapse
|
2
|
Yang Z. The Principle of Cortical Development and Evolution. Neurosci Bull 2025; 41:461-485. [PMID: 39023844 PMCID: PMC11876516 DOI: 10.1007/s12264-024-01259-2] [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: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Human's robust cognitive abilities, including creativity and language, are made possible, at least in large part, by evolutionary changes made to the cerebral cortex. This paper reviews the biology and evolution of mammalian cortical radial glial cells (primary neural stem cells) and introduces the concept that a genetically step wise process, based on a core molecular pathway already in use, is the evolutionary process that has molded cortical neurogenesis. The core mechanism, which has been identified in our recent studies, is the extracellular signal-regulated kinase (ERK)-bone morphogenic protein 7 (BMP7)-GLI3 repressor form (GLI3R)-sonic hedgehog (SHH) positive feedback loop. Additionally, I propose that the molecular basis for cortical evolutionary dwarfism, exemplified by the lissencephalic mouse which originated from a larger gyrencephalic ancestor, is an increase in SHH signaling in radial glia, that antagonizes ERK-BMP7 signaling. Finally, I propose that: (1) SHH signaling is not a key regulator of primate cortical expansion and folding; (2) human cortical radial glial cells do not generate neocortical interneurons; (3) human-specific genes may not be essential for most cortical expansion. I hope this review assists colleagues in the field, guiding research to address gaps in our understanding of cortical development and evolution.
Collapse
Affiliation(s)
- Zhengang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
3
|
Tran LN, Shinde A, Schuster KH, Sabaawy A, Dale E, Welch MJ, Isner TJ, Nunez SA, García-Moreno F, Sagerström CG, Appel BH, Franco SJ. Epigenetic priming of neural progenitors by Notch enhances Sonic hedgehog signaling and establishes gliogenic competence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633996. [PMID: 39896669 PMCID: PMC11785114 DOI: 10.1101/2025.01.20.633996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The remarkable cell diversity of multicellular organisms relies on the ability of multipotent progenitor cells to generate distinct cell types at the right times and locations during embryogenesis. A key question is how progenitors establish competence to respond to the different environmental signals required to produce specific cell types at critical developmental timepoints. We addressed this in the mouse developing forebrain, where neural progenitor cells must switch from producing neurons to making oligodendrocytes in response to increased Sonic Hedgehog (SHH) signaling during late embryogenesis. We show that progenitor responses to SHH are regulated by Notch signaling, thus permitting proper timing of the neuron-oligodendrocyte switch. Notch activity epigenetically primes genes associated with the oligodendrocyte lineage and SHH pathway, enabling amplified transcriptional responses to endogenous SHH and robust oligodendrogenesis. These results reveal a critical role for Notch in facilitating progenitor competence states and influencing cell fate transitions at the epigenetic level.
Collapse
Affiliation(s)
- Luuli N. Tran
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ashwini Shinde
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kristen H. Schuster
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Aiman Sabaawy
- Gates Summer Internship Program, Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Emily Dale
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Madalynn J. Welch
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Trevor J. Isner
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sylvia A. Nunez
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Fernando García-Moreno
- Achucarro Basque Center for Neuroscience, Edificio Sede del Parque Científico de la UPV/EHU, Leioa, Spain
| | - Charles G. Sagerström
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bruce H. Appel
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Gates Summer Internship Program, Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Santos J. Franco
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Gates Summer Internship Program, Gates Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Cell Biology, Stem Cells, and Development Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Program in Pediatric Stem Cell Biology, Children’s Hospital Colorado, Aurora, CO 80045, USA
- Lead contact
| |
Collapse
|
4
|
Hidalgo-Sánchez M, Sánchez-Guardado L, Rodríguez-León J, Francisco-Morcillo J. The role of FGF15/FGF19 in the development of the central nervous system, eyes and inner ears in vertebrates. Tissue Cell 2024; 91:102619. [PMID: 39579736 DOI: 10.1016/j.tice.2024.102619] [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: 07/23/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
Fibroblast growth factor 19 (FGF19), and its rodent ortholog FGF15, is a member of a FGF subfamily directly involved in metabolism, acting in an endocrine way. During embryonic development, FGF15/FGF19 also functions as a paracrine or autocrine factor, regulating key events in a large number of organs. In this sense, the Fgf15/Fgf19 genes control the correct development of the brain, eye, inner ear, heart, pharyngeal pouches, tail bud and limbs, among other organs, as well as muscle growth in adulthood. These growth factors show relevant differences according to molecular structures, signalling pathway and function. Moreover, their expression patterns are highly dynamic at different stages of development, in particular in the central nervous system. The difficulty in understanding the action of these genes increases when comparing their expression patterns and regulatory mechanisms between different groups of vertebrates. The present review will address the expression patterns and functions of the Fgf15/Fgf19 genes at different stages of vertebrate embryonic development, with special attention to the regulation of the early specification, cell differentiation, and morphogenesis of the central nervous system and some sensory organs such as eye and inner ear. The most relevant anatomical aspects related to the structures analysed have also been considered in detail to provide an understandable context for the molecular and cellular studies shown.
Collapse
Affiliation(s)
- Matías Hidalgo-Sánchez
- Área de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain.
| | - Luis Sánchez-Guardado
- Área de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain
| | - Joaquín Rodríguez-León
- Área de Anatomía Humana, Facultad de Medicina y Ciencias de la Salud, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain
| | - Javier Francisco-Morcillo
- Área de Biología Celular, Facultad de Ciencias, Universidad de Extremadura, Avda. de Elvas s/n, Badajoz 06071, Spain
| |
Collapse
|
5
|
Zhang J, Li H, Niswander LA. m 5C methylated lncRncr3-MeCP2 interaction restricts miR124a-initiated neurogenesis. Nat Commun 2024; 15:5136. [PMID: 38879605 PMCID: PMC11180186 DOI: 10.1038/s41467-024-49368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 06/03/2024] [Indexed: 06/19/2024] Open
Abstract
Coordination of neuronal differentiation with expansion of the neuroepithelial/neural progenitor cell (NEPC/NPC) pool is essential in early brain development. Our in vitro and in vivo studies identify independent and opposing roles for two neural-specific and differentially expressed non-coding RNAs derived from the same locus: the evolutionarily conserved lncRNA Rncr3 and the embedded microRNA miR124a-1. Rncr3 regulates NEPC/NPC proliferation and controls the biogenesis of miR124a, which determines neuronal differentiation. Rncr3 conserved exons 2/3 are cytosine methylated and bound by methyl-CpG binding protein MeCP2, which restricts expression of miR124a embedded in exon 4 to prevent premature neuronal differentiation, and to orchestrate proper brain growth. MeCP2 directly binds cytosine-methylated Rncr3 through previously unrecognized lysine residues and suppresses miR124a processing by recruiting PTBP1 to block access of DROSHA-DGCR8. Thus, miRNA processing is controlled by lncRNA m5C methylation along with the defined m5C epitranscriptomic RNA reader protein MeCP2 to coordinate brain development.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Molecular, Cellular, and Developmental Biology. University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - Huili Li
- Department of Molecular, Cellular, and Developmental Biology. University of Colorado Boulder, Boulder, CO, 80309, USA
| | - Lee A Niswander
- Department of Molecular, Cellular, and Developmental Biology. University of Colorado Boulder, Boulder, CO, 80309, USA.
| |
Collapse
|
6
|
Cai E, Barba MG, Ge X. Hedgehog Signaling in Cortical Development. Cells 2023; 13:21. [PMID: 38201225 PMCID: PMC10778342 DOI: 10.3390/cells13010021] [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: 11/15/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
The Hedgehog (Hh) pathway plays a crucial role in embryonic development, acting both as a morphogenic signal that organizes tissue formation and a potent mitogenic signal driving cell proliferation. Dysregulated Hh signaling leads to various developmental defects in the brain. This article aims to review the roles of Hh signaling in the development of the neocortex in the mammalian brain, focusing on its regulation of neural progenitor proliferation and neuronal production. The review will summarize studies on genetic mouse models that have targeted different components of the Hh pathway, such as the ligand Shh, the receptor Ptch1, the GPCR-like transducer Smo, the intracellular transducer Sufu, and the three Gli transcription factors. As key insights into the Hh signaling transduction mechanism were obtained from mouse models displaying neural tube defects, this review will also cover some studies on Hh signaling in neural tube development. The results from these genetic mouse models suggest an intriguing hypothesis that elevated Hh signaling may play a role in the gyrification of the brain in certain species. Additionally, the distinctive production of GABAergic interneurons in the dorsal cortex in the human brain may also be linked to the extension of Hh signaling from the ventral to the dorsal brain region. Overall, these results suggest key roles of Hh signaling as both a morphogenic and mitogenic signal during the forebrain development and imply the potential involvement of Hh signaling in the evolutionary expansion of the neocortex.
Collapse
Affiliation(s)
| | | | - Xuecai Ge
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California Merced, Merced, CA 95340, USA
| |
Collapse
|
7
|
Gayden J, Puig S, Srinivasan C, Phan BN, Abdelhady G, Buck SA, Gamble MC, Tejeda HA, Dong Y, Pfenning AR, Logan RW, Freyberg Z. Integrative multi-dimensional characterization of striatal projection neuron heterogeneity in adult brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.04.539488. [PMID: 37205475 PMCID: PMC10187292 DOI: 10.1101/2023.05.04.539488] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Striatal projection neurons (SPNs) are traditionally segregated into two subpopulations expressing dopamine (DA) D1-like or D2-like receptors. However, this dichotomy is challenged by recent evidence. Functional and expression studies raise important questions: do SPNs co-express different DA receptors, and do these differences reflect unique striatal spatial distributions and expression profiles? Using RNAscope in mouse striatum, we report heterogenous SPN subpopulations distributed across dorsal-ventral and rostral-caudal axes. SPN subpopulations co-express multiple DA receptors, including D1 and D2 (D1/2R) and D1 and D3. Our integrative approach using single-nuclei multi-omics analyses provides a simple consensus to describe SPNs across diverse datasets, connecting it to complementary spatial mapping. Combining RNAscope and multi-omics shows D1/2R SPNs further separate into distinct subtypes according to spatial organization and conserved marker genes. Each SPN cell type contributes uniquely to genetic risk for neuropsychiatric diseases. Our results bridge anatomy and transcriptomics to offer new understandings of striatal neuron heterogeneity.
Collapse
Affiliation(s)
- Jenesis Gayden
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Stephanie Puig
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Chaitanya Srinivasan
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - BaDoi N. Phan
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Medical-Scientist Training Program, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ghada Abdelhady
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Silas A. Buck
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mackenzie C. Gamble
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Molecular and Translational Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Hugo A. Tejeda
- Unit on Neuromodulation and Synaptic Integration, National Institute of Mental Health, Bethesda, MD 20894, USA
| | - Yan Dong
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Andreas R. Pfenning
- Department of Computational Biology, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Ryan W. Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
8
|
Kodera T, Takeuchi RF, Takahashi S, Suzuki K, Kassai H, Aiba A, Shiozawa S, Okano H, Osakada F. Modeling the marmoset brain using embryonic stem cell-derived cerebral assembloids. Biochem Biophys Res Commun 2023; 657:119-127. [PMID: 37002985 DOI: 10.1016/j.bbrc.2023.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023]
Abstract
Studying the non-human primate (NHP) brain is required for the translation of rodent research to humans, but remains a challenge for molecular, cellular, and circuit-level analyses in the NHP brain due to the lack of in vitro NHP brain system. Here, we report an in vitro NHP cerebral model using marmoset (Callithrix jacchus) embryonic stem cell-derived cerebral assembloids (CAs) that recapitulate inhibitory neuron migration and cortical network activity. Cortical organoids (COs) and ganglionic eminence organoids (GEOs) were induced from cjESCs and fused to generate CAs. GEO cells expressing the inhibitory neuron marker LHX6 migrated toward the cortical side of CAs. COs developed their spontaneous neural activity from a synchronized pattern to an unsynchronized pattern as COs matured. CAs containing excitatory and inhibitory neurons showed mature neural activity with an unsynchronized pattern. The CAs represent a powerful in vitro model for studying excitatory and inhibitory neuron interactions, cortical dynamics, and their dysfunction. The marmoset assembloid system will provide an in vitro platform for the NHP neurobiology and facilitate translation into humans in neuroscience research, regenerative medicine, and drug discovery.
Collapse
|
9
|
Cheng B, Sharma DR, Kumar A, Sheth H, Agyemang A, Aschner M, Zhang X, Ballabh P. Shh activation restores interneurons and cognitive function in newborns with intraventricular haemorrhage. Brain 2023; 146:629-644. [PMID: 35867870 PMCID: PMC10169407 DOI: 10.1093/brain/awac271] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/27/2022] [Accepted: 07/05/2022] [Indexed: 11/14/2022] Open
Abstract
Premature infants with germinal matrix haemorrhage-intraventricular haemorrhage (GMH-IVH) suffer from neurobehavioural deficits as they enter childhood and adolescence. Yet the underlying mechanisms remain unclear. Impaired development and function of interneurons contribute to neuropsychiatric disorders. Therefore, we hypothesized that the occurrence of IVH would reduce interneuron neurogenesis in the medial ganglionic eminence and diminish the population of parvalbumin+ and somatostatin+ cortical interneurons. Because Sonic Hedgehog promotes the production of cortical interneurons, we also postulated that the activation of Sonic Hedgehog signalling might restore neurogenesis, cortical interneuron population, and neurobehavioural function in premature newborns with IVH. These hypotheses were tested in a preterm rabbit model of IVH and autopsy samples from human preterm infants. We compared premature newborns with and without IVH for intraneuronal progenitors, cortical interneurons, transcription factors regulating neurogenesis, single-cell transcriptome of medial ganglionic eminence and neurobehavioural functions. We treated premature rabbit kits with adenovirus expressing Sonic Hedgehog (Ad-Shh) or green fluorescence protein gene to determine the effect of Sonic Hedgehog activation on the interneuron production, cortical interneuron population and neurobehaviour. We discovered that IVH reduced the number of Nkx2.1+ and Dlx2+ progenitors in the medial ganglionic eminence of both humans and rabbits by attenuating their proliferation and inducing apoptosis. Moreover, IVH decreased the population of parvalbumin+ and somatostatin+ neurons in the frontal cortex of both preterm infants and kits relative to controls. Sonic Hedgehog expression and the downstream transcription factors, including Nkx2.1, Mash1, Lhx6 and Sox6, were also reduced in kits with IVH. Consistent with these findings, single-cell transcriptomic analyses of medial ganglionic eminence identified a distinct subpopulation of cells exhibiting perturbation in genes regulating neurogenesis, ciliogenesis, mitochondrial function and MAPK signalling in rabbits with IVH. More importantly, restoration of Sonic Hedgehog level by Ad-Shh treatment ameliorated neurogenesis, cortical interneuron population and neurobehavioural function in kits with IVH. Additionally, Sonic Hedgehog activation alleviated IVH-induced inflammation and several transcriptomic changes in the medial ganglionic eminence. Taken together, IVH reduced intraneuronal production and cortical interneuron population by downregulating Sonic Hedgehog signalling in both preterm rabbits and humans. Notably, activation of Sonic Hedgehog signalling restored interneuron neurogenesis, cortical interneurons and cognitive function in rabbit kits with IVH. These findings highlight disruption in cortical interneurons in IVH and identify a novel therapeutic strategy to restore cortical interneurons and cognitive function in infants with IVH. These studies can accelerate the development of new therapies to enhance the neurodevelopmental outcome of survivors with IVH.
Collapse
Affiliation(s)
- Bokun Cheng
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deep R Sharma
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ajeet Kumar
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hardik Sheth
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alex Agyemang
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Aschner
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xusheng Zhang
- Computational Genomics Core, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
10
|
Ni P, Fan L, Zinski A, Chung S. Generation of Homogeneous Populations of Cortical Interneurons from Human Pluripotent Stem Cells. Methods Mol Biol 2023; 2683:13-20. [PMID: 37300763 DOI: 10.1007/978-1-0716-3287-1_2] [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] [Indexed: 06/12/2023]
Abstract
Cortical interneurons (cINs), especially those that are derived from the medial ganglionic eminence (MGE) during early development, are associated with various neuropsychiatric disorders. Human pluripotent stem cell (hPSC)-derived cINs can provide unlimited cell sources for studying disease mechanisms and developing novel therapeutics. Here, we describe an optimized method to generate homogeneous cIN populations based on three-dimensional (3D) cIN sphere generation. This optimized differentiation system could sustain generated cINs relatively long term without compromising their survival or phenotypes.
Collapse
Affiliation(s)
- Peiyan Ni
- Psychiatric Laboratory and Mental Health Center, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyi Fan
- Psychiatric Laboratory and Mental Health Center, The State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Amy Zinski
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Sangmi Chung
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA.
| |
Collapse
|
11
|
Gupta R, Mehan S, Chhabra S, Giri A, Sherawat K. Role of Sonic Hedgehog Signaling Activation in the Prevention of Neurological Abnormalities Associated with Obsessive-Compulsive Disorder. Neurotox Res 2022; 40:1718-1738. [PMID: 36272053 DOI: 10.1007/s12640-022-00586-4] [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: 04/02/2022] [Revised: 09/15/2022] [Accepted: 10/07/2022] [Indexed: 12/31/2022]
Abstract
The smoothened sonic hedgehog (Smo-Shh) pathway is one mechanism that influences neurogenesis, including brain cell differentiation and development during childhood. Shh signaling dysregulation leads to decreased target gene transcription, which contributes to increased neuronal excitation, apoptosis, and neurodegeneration, eventually leading to neurological deficits. Neuropsychiatric disorders such as OCD and related neurological dysfunctions are characterized by neurotransmitter imbalance, neuroinflammation, oxidative stress, and impaired neurogenesis, disturbing the cortico-striato-thalamo-cortical (CSTC) link neuronal network. Despite the availability of several treatments, such as selective serotonin reuptake inhibitors, some individuals may not benefit much from them. Several trials on the use of antipsychotics in the treatment of OCD have also produced inadequate findings. This evidence-based review focuses on a potential pharmacological approach to alleviating OCD and associated neuronal deficits by preventing neurochemical alterations, in which sonic hedgehog activators are neuroprotective, lowering neuronal damage while increasing neuronal maintenance and survival. As a result, stimulating SMO-Shh via its potential activators may have neuroprotective effects on neurological impairment associated with OCD. This review investigates the link between SMO-Shh signaling and the neurochemical abnormalities associated with the progression of OCD and associated neurological dysfunctions. Role of Smo-Shh signaling in serotonergic neurogenesis and in maintaining their neuronal identity. The Shh ligand activates two main transcriptional factors known as Foxa2 and Nkx2.2, which again activates another transcriptional factor, GATA (GATA2 and GATA3), in post mitotic precursor cells of serotonergic neurons-following increased expression of Pet-1 and Lmx1b after GATA regulates the expression of many serotonergic enzymes such as TPH2, SERT, VMAT, slc6a4, Htr1a, Htr1b (Serotonin receptor enzymes), and MAO that regulate and control the release of serotonin and maintain their neuronal identity after their maturation. Abbreviation: Foxa2: Forkhead box; GATA: Globin transcription factor; Lmx1b: LIM homeobox transcription factor 1 beta; TPH2: Tryptophan hydroxylase 2; Htr1a: Serotonin receptor 1a; Htr1b: Serotonin receptor 1b; SERT: Serotonin transporter; VMAT: Vesicular monoamine transporter; MAO: Monoamine oxidase.
Collapse
Affiliation(s)
- Ria Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| | - Swesha Chhabra
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Aditi Giri
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Kajal Sherawat
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India
| |
Collapse
|
12
|
Hirose T, Sugitani Y, Kurihara H, Kazama H, Kusaka C, Noda T, Takahashi H, Ohno S. PAR3 restricts the expansion of neural precursor cells by regulating hedgehog signaling. Development 2022; 149:277212. [DOI: 10.1242/dev.199931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/03/2022] [Indexed: 11/07/2022]
Abstract
ABSTRACT
During brain development, neural precursor cells (NPCs) expand initially, and then switch to generating stage-specific neurons while maintaining self-renewal ability. Because the NPC pool at the onset of neurogenesis crucially affects the final number of each type of neuron, tight regulation is necessary for the transitional timing from the expansion to the neurogenic phase in these cells. However, the molecular mechanisms underlying this transition are poorly understood. Here, we report that the telencephalon-specific loss of PAR3 before the start of neurogenesis leads to increased NPC proliferation at the expense of neurogenesis, resulting in disorganized tissue architecture. These NPCs demonstrate hyperactivation of hedgehog signaling in a smoothened-dependent manner, as well as defects in primary cilia. Furthermore, loss of PAR3 enhanced ligand-independent ciliary accumulation of smoothened and an inhibitor of smoothened ameliorated the hyperproliferation of NPCs in the telencephalon. Thus, these findings support the idea that PAR3 has a crucial role in the transition of NPCs from the expansion phase to the neurogenic phase by restricting hedgehog signaling through the establishment of ciliary integrity.
Collapse
Affiliation(s)
- Tomonori Hirose
- Yokohama City University School of Medicine 1 Department of Molecular Biology , , Yokohama 236-0004 , Japan
- Cancer Institute 2 Department of Cell Biology , , , Tokyo 135-8550 , Japan
- Japanese Foundation for Cancer Research 2 Department of Cell Biology , , , Tokyo 135-8550 , Japan
| | - Yoshinobu Sugitani
- Cancer Institute 2 Department of Cell Biology , , , Tokyo 135-8550 , Japan
- Japanese Foundation for Cancer Research 2 Department of Cell Biology , , , Tokyo 135-8550 , Japan
- Juntendo University School of Medicine 3 Department of Pathology and Oncology , , Tokyo 113-8421 , Japan
| | - Hidetake Kurihara
- Juntendo University Graduate School of Medicine 4 Department of Anatomy and Life Structure , , Tokyo 113-8421 , Japan
- Department of Physical Therapy, Faculty of Health Science, Aino University 5 , Osaka 567-0012 , Japan
| | - Hiromi Kazama
- Yokohama City University School of Medicine 1 Department of Molecular Biology , , Yokohama 236-0004 , Japan
| | - Chiho Kusaka
- Yokohama City University School of Medicine 1 Department of Molecular Biology , , Yokohama 236-0004 , Japan
| | - Tetsuo Noda
- Cancer Institute 2 Department of Cell Biology , , , Tokyo 135-8550 , Japan
- Japanese Foundation for Cancer Research 2 Department of Cell Biology , , , Tokyo 135-8550 , Japan
- Director's Room, Cancer Institute, Japanese Foundation for Cancer Research 6 , Tokyo 135-8550 , Japan
| | - Hidehisa Takahashi
- Yokohama City University School of Medicine 1 Department of Molecular Biology , , Yokohama 236-0004 , Japan
| | - Shigeo Ohno
- Yokohama City University School of Medicine 1 Department of Molecular Biology , , Yokohama 236-0004 , Japan
| |
Collapse
|
13
|
Varga BV, Faiz M, Pivonkova H, Khelifi G, Yang H, Gao S, Linderoth E, Zhen M, Karadottir RT, Hussein SM, Nagy A. Signal requirement for cortical potential of transplantable human neuroepithelial stem cells. Nat Commun 2022; 13:2844. [PMID: 35606347 PMCID: PMC9126949 DOI: 10.1038/s41467-022-29839-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/21/2022] [Indexed: 01/26/2023] Open
Abstract
The cerebral cortex develops from dorsal forebrain neuroepithelial progenitor cells. Following the initial expansion of the progenitor cell pool, these cells generate neurons of all the cortical layers and then astrocytes and oligodendrocytes. Yet, the regulatory pathways that control the expansion and maintenance of the progenitor cell pool are currently unknown. Here we define six basic pathway components that regulate proliferation of cortically specified human neuroepithelial stem cells (cNESCs) in vitro without the loss of cerebral cortex developmental potential. We show that activation of FGF and inhibition of BMP and ACTIVIN A signalling are required for long-term cNESC proliferation. We also demonstrate that cNESCs preserve dorsal telencephalon-specific potential when GSK3, AKT and nuclear CATENIN-β1 activity are low. Remarkably, regulation of these six pathway components supports the clonal expansion of cNESCs. Moreover, cNESCs differentiate into lower- and upper-layer cortical neurons in vitro and in vivo. The identification of mechanisms that drive the neuroepithelial stem cell self-renewal and differentiation and preserve this potential in vitro is key to developing regenerative and cell-based therapeutic approaches to treat neurological conditions.
Collapse
Affiliation(s)
- Balazs V Varga
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada. .,Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, UK.
| | - Maryam Faiz
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Surgery, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Helena Pivonkova
- Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, UK
| | - Gabriel Khelifi
- Cancer Research Center, Université Laval, Quebec City, QC, Canada.,CHU of Québec-Université Laval Research Center, Oncology Division, Quebec City, QC, Canada
| | - Huijuan Yang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Shangbang Gao
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Emma Linderoth
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Mei Zhen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ragnhildur Thora Karadottir
- Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Puddicombe Way, Cambridge, UK.,Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Samer M Hussein
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.,Cancer Research Center, Université Laval, Quebec City, QC, Canada.,CHU of Québec-Université Laval Research Center, Oncology Division, Quebec City, QC, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada. .,Department of Obstetrics and Gynaecology, and Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
van Bruggen D, Pohl F, Langseth CM, Kukanja P, Lee H, Albiach AM, Kabbe M, Meijer M, Linnarsson S, Hilscher MM, Nilsson M, Sundström E, Castelo-Branco G. Developmental landscape of human forebrain at a single-cell level identifies early waves of oligodendrogenesis. Dev Cell 2022; 57:1421-1436.e5. [PMID: 35523173 DOI: 10.1016/j.devcel.2022.04.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 03/20/2022] [Accepted: 04/14/2022] [Indexed: 01/06/2023]
Abstract
Oligodendrogenesis in the human central nervous system has been observed mainly at the second trimester of gestation, a much later developmental stage compared to oligodendrogenesis in mice. Here, we characterize the transcriptomic neural diversity in the human forebrain at post-conception weeks (PCW) 8-10. Using single-cell RNA sequencing, we find evidence of the emergence of a first wave of oligodendrocyte lineage cells as early as PCW 8, which we also confirm at the epigenomic level through the use of single-cell ATAC-seq. Using regulatory network inference, we predict key transcriptional events leading to the specification of oligodendrocyte precursor cells (OPCs). Moreover, by profiling the spatial expression of 50 key genes through the use of in situ sequencing (ISS), we identify regions in the human ventral fetal forebrain where oligodendrogenesis first occurs. Our results indicate evolutionary conservation of the first wave of oligodendrogenesis between mice and humans and describe regulatory mechanisms involved in human OPC specification.
Collapse
Affiliation(s)
- David van Bruggen
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Fabio Pohl
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | | | - Petra Kukanja
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Hower Lee
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Alejandro Mossi Albiach
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Mukund Kabbe
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Mandy Meijer
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Sten Linnarsson
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden
| | - Markus M Hilscher
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65, Solna, Sweden
| | - Erik Sundström
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, BioClinicum, Solna, Sweden
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, 17177 Stockholm, Sweden; Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
15
|
Eto H, Kishi Y. Brain regionalization by Polycomb-group proteins and chromatin accessibility. Bioessays 2021; 43:e2100155. [PMID: 34536032 DOI: 10.1002/bies.202100155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/30/2021] [Accepted: 08/30/2021] [Indexed: 11/10/2022]
Abstract
During brain development, neural precursor cells (NPCs) in different brain regions produce different types of neurons, and each of these regions plays a different role in the adult brain. Therefore, precise regionalization is essential in the early stages of brain development, and irregular regionalization has been proposed as the cause of neurodevelopmental disorders. The mechanisms underlying brain regionalization have been well studied in terms of morphogen-induced expression of critical transcription factors for regionalization. NPC potential in different brain regions is defined by chromatin structures that regulate the plasticity of gene expression. Herein, we present recent findings on the importance of chromatin structure in brain regionalization, particularly with respect to its regulation by Polycomb-group proteins and chromatin accessibility.
Collapse
Affiliation(s)
- Hikaru Eto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yusuke Kishi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Meurer L, Ferdman L, Belcher B, Camarata T. The SIX Family of Transcription Factors: Common Themes Integrating Developmental and Cancer Biology. Front Cell Dev Biol 2021; 9:707854. [PMID: 34490256 PMCID: PMC8417317 DOI: 10.3389/fcell.2021.707854] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 01/19/2023] Open
Abstract
The sine oculis (SIX) family of transcription factors are key regulators of developmental processes during embryogenesis. Members of this family control gene expression to promote self-renewal of progenitor cell populations and govern mechanisms of cell differentiation. When the function of SIX genes becomes disrupted, distinct congenital defects develops both in animal models and humans. In addition to the embryonic setting, members of the SIX family have been found to be critical regulators of tumorigenesis, promoting cell proliferation, epithelial-to-mesenchymal transition, and metastasis. Research in both the fields of developmental biology and cancer research have provided an extensive understanding of SIX family transcription factor functions. Here we review recent progress in elucidating the role of SIX family genes in congenital disease as well as in the promotion of cancer. Common themes arise when comparing SIX transcription factor function during embryonic and cancer development. We highlight the complementary nature of these two fields and how knowledge in one area can open new aspects of experimentation in the other.
Collapse
Affiliation(s)
- Logan Meurer
- Department of Basic Sciences, NYIT College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, United States
| | - Leonard Ferdman
- Department of Basic Sciences, NYIT College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, United States
| | - Beau Belcher
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR, United States
| | - Troy Camarata
- Department of Basic Sciences, NYIT College of Osteopathic Medicine at Arkansas State University, Jonesboro, AR, United States
| |
Collapse
|
17
|
Samoilova EM, Belopasov VV, Baklaushev VP. Transcription Factors of Direct Neuronal Reprogramming in Ontogenesis and Ex Vivo. Mol Biol 2021; 55:645-669. [DOI: 10.1134/s0026893321040087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 03/07/2025]
|
18
|
Benito-Gutiérrez È, Gattoni G, Stemmer M, Rohr SD, Schuhmacher LN, Tang J, Marconi A, Jékely G, Arendt D. The dorsoanterior brain of adult amphioxus shares similarities in expression profile and neuronal composition with the vertebrate telencephalon. BMC Biol 2021; 19:110. [PMID: 34020648 PMCID: PMC8139002 DOI: 10.1186/s12915-021-01045-w] [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: 09/18/2020] [Accepted: 05/06/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The evolutionary origin of the telencephalon, the most anterior part of the vertebrate brain, remains obscure. Since no obvious counterpart to the telencephalon has yet been identified in invertebrate chordates, it is difficult to trace telencephalic origins. One way to identify homologous brain parts between distantly related animal groups is to focus on the combinatorial expression of conserved regionalisation genes that specify brain regions. RESULTS Here, we report the combined expression of conserved transcription factors known to specify the telencephalon in the vertebrates in the chordate amphioxus. Focusing on adult specimens, we detect specific co-expression of these factors in the dorsal part of the anterior brain vesicle, which we refer to as Pars anterodorsalis (PAD). As in vertebrates, expression of the transcription factors FoxG1, Emx and Lhx2/9 overlaps that of Pax4/6 dorsally and of Nkx2.1 ventrally, where we also detect expression of the Hedgehog ligand. This specific pattern of co-expression is not observed prior to metamorphosis. Similar to the vertebrate telencephalon, the amphioxus PAD is characterised by the presence of GABAergic neurons and dorsal accumulations of glutamatergic as well as dopaminergic neurons. We also observe sustained proliferation of neuronal progenitors at the ventricular zone of the amphioxus brain vesicle, as observed in the vertebrate brain. CONCLUSIONS Our findings suggest that the PAD in the adult amphioxus brain vesicle and the vertebrate telencephalon evolved from the same brain precursor region in ancestral chordates, which would imply homology of these structures. Our comparative data also indicate that this ancestral brain already contained GABA-, glutamatergic and dopaminergic neurons, as is characteristic for the olfactory bulb of the vertebrate telencephalon. We further speculate that the telencephalon might have evolved in vertebrates via a heterochronic shift in developmental timing.
Collapse
Affiliation(s)
- Èlia Benito-Gutiérrez
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK.
| | - Giacomo Gattoni
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Manuel Stemmer
- Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
- Present Address: Max-Planck Institute for Neurobiology in Martinsried, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Silvia D Rohr
- Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Laura N Schuhmacher
- Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany
- Present Address: Department of Cell & Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jocelyn Tang
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Aleksandra Marconi
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | - Gáspár Jékely
- Living Systems Institute, University of Exeter, Exeter, EX4 4QD, UK
| | - Detlev Arendt
- Developmental Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| |
Collapse
|
19
|
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
|
20
|
Wen Y, Su Z, Wang Z, Yang L, Liu G, Shang Z, Duan Y, Du H, Li Z, You Y, Li X, Yang Z, Zhang Z. Transcription Factor VAX1 Regulates the Regional Specification of the Subpallium Through Repressing Gsx2. Mol Neurobiol 2021; 58:3729-3744. [PMID: 33821423 DOI: 10.1007/s12035-021-02378-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/29/2021] [Indexed: 12/22/2022]
Abstract
Specification of the progenitors' regional identity is a pivotal step during development of the cerebral cortex and basal ganglia. The molecular mechanisms underlying progenitor regionalization, however, are poorly understood. Here we showed that the transcription factor Vax1 was highly expressed in the developing subpallium. In its absence, the RNA-Seq analysis, in situ RNA hybridization, and immunofluorescence staining results showed that the cell proliferation was increased in the subpallium, but the neuronal differentiation was blocked. Moreover, the dLGE expands ventrally, and the vLGE, MGE, and septum get smaller. Finally, overexpressed VAX1 in the LGE progenitors strongly inhibits Gsx2 expression. Taken together, our findings show that Vax1 is crucial for subpallium regionalization by repressing Gsx2.
Collapse
Affiliation(s)
- Yan Wen
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Zihao Su
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Ziwu Wang
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Lin Yang
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Guoping Liu
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Zicong Shang
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Yangyang Duan
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Heng Du
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Zhenmeiyu Li
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Yan You
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Xiaosu Li
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Zhengang Yang
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Zhuangzhi Zhang
- Department of Neurology, Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China.
| |
Collapse
|
21
|
Abstract
Brain organoids closely recapitulate many features and characteristics of in vivo brain tissue. This technology in turn allows unprecedented possibilities to investigate brain development and function in the dish. Several brain organoid protocols have been established, and the studies have focused on validating the architecture, cellular composition, and function of the organoids. In future, the improved and advanced organoid models will enable us to understand cellular and molecular features of the developing brain. However, several obstacles, such as the quality of the organoids, 3D structural analysis, and measurement of the neural connectivity need to be improved. In this perspective, we will provide an overview of the current state of the art of the brain organoid field, with a focus on protocols and organoid characterization. Additionally, we will address the current limitations of this evolving field and provide an understanding of the current brain organoid landscape and insight toward the next steps.
Collapse
|
22
|
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
|
23
|
Willsey HR, Exner CRT, Xu Y, Everitt A, Sun N, Wang B, Dea J, Schmunk G, Zaltsman Y, Teerikorpi N, Kim A, Anderson AS, Shin D, Seyler M, Nowakowski TJ, Harland RM, Willsey AJ, State MW. Parallel in vivo analysis of large-effect autism genes implicates cortical neurogenesis and estrogen in risk and resilience. Neuron 2021; 109:788-804.e8. [PMID: 33497602 DOI: 10.1016/j.neuron.2021.01.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/02/2020] [Accepted: 01/04/2021] [Indexed: 12/29/2022]
Abstract
Gene Ontology analyses of autism spectrum disorders (ASD) risk genes have repeatedly highlighted synaptic function and transcriptional regulation as key points of convergence. However, these analyses rely on incomplete knowledge of gene function across brain development. Here we leverage Xenopus tropicalis to study in vivo ten genes with the strongest statistical evidence for association with ASD. All genes are expressed in developing telencephalon at time points mapping to human mid-prenatal development, and mutations lead to an increase in the ratio of neural progenitor cells to maturing neurons, supporting previous in silico systems biological findings implicating cortical neurons in ASD vulnerability, but expanding the range of convergent functions to include neurogenesis. Systematic chemical screening identifies that estrogen, via Sonic hedgehog signaling, rescues this convergent phenotype in Xenopus and human models of brain development, suggesting a resilience factor that may mitigate a range of ASD genetic risks.
Collapse
Affiliation(s)
- Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cameron R T Exner
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yuxiao Xu
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Amanda Everitt
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nawei Sun
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Belinda Wang
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jeanselle Dea
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Galina Schmunk
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yefim Zaltsman
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nia Teerikorpi
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Albert Kim
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Aoife S Anderson
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David Shin
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Meghan Seyler
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tomasz J Nowakowski
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Richard M Harland
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - A Jeremy Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Institute for Neurodegenerative Diseases, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Matthew W State
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94143, USA; Langley Porter Psychiatric Institute, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
24
|
Eto H, Kishi Y, Yakushiji-Kaminatsui N, Sugishita H, Utsunomiya S, Koseki H, Gotoh Y. The Polycomb group protein Ring1 regulates dorsoventral patterning of the mouse telencephalon. Nat Commun 2020; 11:5709. [PMID: 33177537 PMCID: PMC7658352 DOI: 10.1038/s41467-020-19556-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 10/16/2020] [Indexed: 12/30/2022] Open
Abstract
Dorsal-ventral patterning of the mammalian telencephalon is fundamental to the formation of distinct functional regions including the neocortex and ganglionic eminence. While Bone morphogenetic protein (BMP), Wnt, and Sonic hedgehog (Shh) signaling are known to determine regional identity along the dorsoventral axis, how the region-specific expression of these morphogens is established remains unclear. Here we show that the Polycomb group (PcG) protein Ring1 contributes to the ventralization of the mouse telencephalon. Deletion of Ring1b or both Ring1a and Ring1b in neuroepithelial cells induces ectopic expression of dorsal genes, including those for BMP and Wnt ligands, as well as attenuated expression of the gene for Shh, a key morphogen for ventralization, in the ventral telencephalon. We observe PcG protein–mediated trimethylation of histone 3 at lysine-27 and binding of Ring1B at BMP and Wnt ligand genes specifically in the ventral region. Furthermore, forced activation of BMP or Wnt signaling represses Shh expression. Our results thus indicate that PcG proteins suppress BMP and Wnt signaling in a region-specific manner and thereby allow proper Shh expression and development of the ventral telencephalon. NCOMMS-19-38235B Dorsal-ventral patterning of the mammalian telencephalon is fundamental to the formation of distinct functional regions. Here, the authors find that PcG proteins suppress BMP and Wnt signaling in a region-specific manner, allowing for proper Shh expression and development of the ventral telencephalon.
Collapse
Affiliation(s)
- Hikaru Eto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yusuke Kishi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Nayuta Yakushiji-Kaminatsui
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Hiroki Sugishita
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Shun Utsunomiya
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyoku, Tokyo, 113-0033, Japan.,Neuroscience 2, Drug Discovery & Disease Research Laboratory, Shionogi & Co., Ltd.; Business-Academia Collaborative Laboratory (Shionogi), Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (RIKEN-IMS), 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Yukiko Gotoh
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
25
|
Du A, Wu X, Chen H, Bai QR, Han X, Liu B, Zhang X, Ding Z, Shen Q, Zhao C. Foxg1 Directly Represses Dbx1 to Confine the POA and Subsequently Regulate Ventral Telencephalic Patterning. Cereb Cortex 2020; 29:4968-4981. [PMID: 30843579 DOI: 10.1093/cercor/bhz037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/01/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
During early development, signaling centers, such as the cortical hem and the preoptic area (POA), are critical for telencephalic patterning. However, the mechanisms underlying the maintenance of signal centers are poorly understood. Here, we report that the transcription factor Foxg1 is required to confine the POA, a resource of Sonic Hedgehog (Shh) that is pivotal for ventral telencephalic development. Cell-specific deletion of Foxg1 achieved by crossing Foxg1fl/fl with Dbx1-cre or Nestin-CreER combined with tamoxifen induction results in a dramatic expansion of the POA accompanied by the significantly increased activity of the Shh signaling pathway. Ventral pattern formation was severely impaired. Moreover, we demonstrated that Foxg1 directly represses Dbx1 to restrict the POA. Furthermore, we found that the ventral pallium was expanded, which might also contribute to the observed patterning defects. These findings will improve our understanding of the maintenance of signal centers and help to elucidate the mechanisms underlying ventral telencephalic patterning.
Collapse
Affiliation(s)
- Ailing Du
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaojing Wu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Hanhan Chen
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qing-Ran Bai
- Tongji Hospital, Brain and Spinal Cord Innovative Research Center, School of Life Sciences and Technology, Tongji University, Shanghai 200065, China
| | - Xiao Han
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bin Liu
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xiaohu Zhang
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Zhaoying Ding
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Qin Shen
- Tongji Hospital, Brain and Spinal Cord Innovative Research Center, School of Life Sciences and Technology, Tongji University, Shanghai 200065, China
| | - Chunjie Zhao
- Key Laboratory of Developmental Genes and Human Diseases, MOE, Department of histology and embryology, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
26
|
Göngrich C, Krapacher FA, Munguba H, Fernández-Suárez D, Andersson A, Hjerling-Leffler J, Ibáñez CF. ALK4 coordinates extracellular and intrinsic signals to regulate development of cortical somatostatin interneurons. J Cell Biol 2020; 219:jcb.201905002. [PMID: 31676717 PMCID: PMC7039195 DOI: 10.1083/jcb.201905002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/03/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Göngrich et al. show that the activin receptor ALK4 is a key regulator of the specification of somatostatin interneurons. They find that intrinsic transcriptional programs interact with extracellular signals present in the environment of MGE cells to regulate cortical interneuron specification. Although the role of transcription factors in fate specification of cortical interneurons is well established, how these interact with extracellular signals to regulate interneuron development is poorly understood. Here we show that the activin receptor ALK4 is a key regulator of the specification of somatostatin interneurons. Mice lacking ALK4 in GABAergic neurons of the medial ganglionic eminence (MGE) showed marked deficits in distinct subpopulations of somatostatin interneurons from early postnatal stages of cortical development. Specific losses were observed among distinct subtypes of somatostatin+/Reelin+ double-positive cells, including Hpse+ layer IV cells targeting parvalbumin+ interneurons, leading to quantitative alterations in the inhibitory circuitry of this layer. Activin-mediated ALK4 signaling in MGE cells induced interaction of Smad2 with SATB1, a transcription factor critical for somatostatin interneuron development, and promoted SATB1 nuclear translocation and repositioning within the somatostatin gene promoter. These results indicate that intrinsic transcriptional programs interact with extracellular signals present in the environment of MGE cells to regulate cortical interneuron specification.
Collapse
Affiliation(s)
| | | | - Hermany Munguba
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | | | - Annika Andersson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Carlos F Ibáñez
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden.,Department of Physiology, National University of Singapore, Singapore.,Life Sciences Institute, National University of Singapore, Singapore.,Stellenbosch Institute for Advanced Study, Wallenberg Research Centre at Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
27
|
Delgado RN, Mansky B, Ahanger SH, Lu C, Andersen RE, Dou Y, Alvarez-Buylla A, Lim DA. Maintenance of neural stem cell positional identity by mixed-lineage leukemia 1. Science 2020; 368:48-53. [PMID: 32241942 DOI: 10.1126/science.aba5960] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Neural stem cells (NSCs) in the developing and postnatal brain have distinct positional identities that dictate the types of neurons they generate. Although morphogens initially establish NSC positional identity in the neural tube, it is unclear how such regional differences are maintained as the forebrain grows much larger and more anatomically complex. We found that the maintenance of NSC positional identity in the murine brain requires a mixed-lineage leukemia 1 (Mll1)-dependent epigenetic memory system. After establishment by sonic hedgehog, ventral NSC identity became independent of this morphogen. Even transient MLL1 inhibition caused a durable loss of ventral identity, resulting in the generation of neurons with the characteristics of dorsal NSCs in vivo. Thus, spatial information provided by morphogens can be transitioned to epigenetic mechanisms that maintain regionally distinct developmental programs in the forebrain.
Collapse
Affiliation(s)
- Ryan N Delgado
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Benjamin Mansky
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sajad Hamid Ahanger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Changqing Lu
- Department of Human Anatomy, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Rebecca E Andersen
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yali Dou
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| |
Collapse
|
28
|
Ma M, Legué E, Tian X, Somlo S, Liem KF. Cell-Autonomous Hedgehog Signaling Is Not Required for Cyst Formation in Autosomal Dominant Polycystic Kidney Disease. J Am Soc Nephrol 2019; 30:2103-2111. [PMID: 31451534 DOI: 10.1681/asn.2018121274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/15/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND PKD1 or PKD2, the two main causal genes for autosomal dominant polycystic kidney disease (ADPKD), encode the multipass transmembrane proteins polycystin-1 (PC1) and polycystin-2 (PC2), respectively. Polycystins localize to the primary cilium, an organelle essential for cell signaling, including signal transduction of the Hedgehog pathway. Mutations in ciliary genes that build and maintain the cilium also cause renal cystic disease through unknown pathways. Although recent studies have found alterations in Hedgehog signaling in ADPKD-related models and tissues, the relationship between Hedgehog and polycystic kidney disease is not known. METHODS To examine the potential role of cell-autonomous Hedgehog signaling in regulating kidney cyst formation in vivo in both early- and adult-onset mouse models of ADPKD, we used conditional inactivation of Pkd1 combined with conditional modulation of Hedgehog signaling components in renal epithelial cells, where mutations in Pkd1 initiate cyst formation. After increasing or decreasing levels of Hedgehog signaling in cells that underwent inactivation of Pkd1, we evaluated the effects of these genetic manipulations on quantitative parameters of polycystic kidney disease severity. RESULTS We found that in Pkd1 conditional mutant mouse kidneys, neither downregulation nor activation of the Hedgehog pathway in epithelial cells along the nephron significantly influenced the severity of the polycystic kidney phenotype in mouse models of developmental or adult-onset of ADPKD. CONCLUSIONS These data suggest that loss of Pkd1 function results in kidney cysts through pathways that are not affected by the activity of the Hedgehog pathway.
Collapse
Affiliation(s)
- Ming Ma
- Departments of Internal Medicine
| | - Emilie Legué
- Pediatrics, and.,Vertebrate Developmental Biology Program, Yale University, New Haven, Connecticut
| | - Xin Tian
- Departments of Internal Medicine
| | | | - Karel F Liem
- Pediatrics, and .,Vertebrate Developmental Biology Program, Yale University, New Haven, Connecticut
| |
Collapse
|
29
|
Winkler CC, Franco SJ. Loss of Shh signaling in the neocortex reveals heterogeneous cell recovery responses from distinct oligodendrocyte populations. Dev Biol 2019; 452:55-65. [PMID: 31071314 DOI: 10.1016/j.ydbio.2019.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/25/2019] [Accepted: 04/25/2019] [Indexed: 10/26/2022]
Abstract
The majority of oligodendrocytes in the neocortex originate from neural progenitors that reside in the dorsal forebrain. We recently showed that Sonic Hedgehog (Shh) signaling in these dorsal progenitors is required to produce normal numbers of neocortical oligodendrocytes during embryonic development. Conditional deletion of the Shh signaling effector, Smo, in dorsal progenitors caused a dramatic reduction in oligodendrocyte numbers in the embryonic neocortex. In the current study, we show that the depleted oligodendrocyte lineage in Smo conditional mutants is able to recover to control numbers over time. This eventual recovery is achieved in part by expansion of the ventrally-derived wild-type lineage that normally makes up a minority of the total oligodendrocyte population. However, we find that the remaining dorsally-derived mutant cells also increase in numbers over time to contribute equally to the recovery of the total population. Additionally, we found that the ways in which the dorsal and ventral sources cooperate to achieve recovery is different for distinct populations of oligodendrocyte-lineage cells. Oligodendrocyte precursor cells (OPCs) in the neocortical white matter recover completely by expansion of the remaining dorsally-derived Smo mutant cells. On the other hand, mature oligodendrocytes in the white and gray matter recover through an equal contribution from dorsal mutant and ventral wild-type lineages. Interestingly, the only population that did not make a full recovery was OPCs in the gray matter. We find that gray matter OPCs are less proliferative in Smo cKO mutants compared to controls, which may explain their inability to fully recover. Our data indicate that certain populations of the dorsal oligodendrocyte lineage are more affected by loss of Shh signaling than others. Furthermore, these studies shed new light on the complex relationship between dorsal and ventral sources of oligodendrocytes in the developing neocortex.
Collapse
Affiliation(s)
- Caitlin C Winkler
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA; Cell Biology, Stem Cells and Development Graduate Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Santos J Franco
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA; Cell Biology, Stem Cells and Development Graduate Program, University of Colorado School of Medicine, Aurora, CO, 80045, USA; Program of Pediatric Stem Cell Biology, Children's Hospital Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
30
|
Rushing GV, Bollig MK, Ihrie RA. Heterogeneity of Neural Stem Cells in the Ventricular-Subventricular Zone. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1169:1-30. [PMID: 31487016 DOI: 10.1007/978-3-030-24108-7_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this chapter, heterogeneity is explored in the context of the ventricular-subventricular zone, the largest stem cell niche in the mammalian brain. This niche generates up to 10,000 new neurons daily in adult mice and extends over a large spatial area with dorso-ventral and medio-lateral subdivisions. The stem cells of the ventricular-subventricular zone can be subdivided by their anatomical position and transcriptional profile, and the stem cell lineage can also be further subdivided into stages of pre- and post-natal quiescence and activation. Beyond the stem cells proper, additional differences exist in their interactions with other cellular constituents of the niche, including neurons, vasculature, and cerebrospinal fluid. These variations in stem cell potential and local interactions are discussed, as well as unanswered questions within this system.
Collapse
Affiliation(s)
- Gabrielle V Rushing
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Neuroscience Program, Vanderbilt University, Nashville, TN, USA
| | - Madelyn K Bollig
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.,Neuroscience Program, Vanderbilt University, Nashville, TN, USA
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Neuroscience Program, Vanderbilt University, Nashville, TN, USA. .,Department of Neurological Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
31
|
The Dorsal Wave of Neocortical Oligodendrogenesis Begins Embryonically and Requires Multiple Sources of Sonic Hedgehog. J Neurosci 2018; 38:5237-5250. [PMID: 29739868 DOI: 10.1523/jneurosci.3392-17.2018] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 01/06/2023] Open
Abstract
Neural progenitor cells in the developing dorsal forebrain give rise to excitatory neurons, astrocytes, and oligodendrocytes for the neocortex. While we are starting to gain a better understanding about the mechanisms that direct the formation of neocortical neurons and astrocytes, far less is known about the molecular mechanisms that instruct dorsal forebrain progenitors to make oligodendrocytes. In this study, we show that Sonic hedgehog (Shh) signaling is required in dorsal progenitors for their late embryonic transition to oligodendrogenesis. Using genetic lineage-tracing in mice of both sexes, we demonstrate that most oligodendrocytes in the embryonic neocortex derive from Emx1+ dorsal forebrain progenitors. Deletion of the Shh signaling effector Smo specifically in Emx1+ progenitors led to significantly decreased oligodendrocyte numbers in the embryonic neocortex. Conversely, knock-out of the Shh antagonist Sufu was sufficient to increase neocortical oligodendrogenesis. Using conditional knock-out strategies, we found that Shh ligand is supplied to dorsal progenitors through multiple sources. Loss of Shh from Dlx5/6+ interneurons caused a significant reduction in oligodendrocytes in the embryonic neocortex. This phenotype was identical to that observed upon Shh deletion from the entire CNS using Nestin-Cre, indicating that interneurons migrating into the neocortex from the subpallium are the primary neural source of Shh for dorsal oligodendrogenesis. Additionally, deletion of Shh from migrating interneurons together with the choroid plexus epithelium led to a more severe loss of oligodendrocytes, suggesting that the choroid plexus is an important non-neural source of Shh ligand. Together, our studies demonstrate that the dorsal wave of neocortical oligodendrogenesis occurs earlier than previously appreciated and requires highly regulated Shh signaling from multiple embryonic sources.SIGNIFICANCE STATEMENT Most neocortical oligodendrocytes are made by neural progenitors in the dorsal forebrain, but the mechanisms that specify this fate are poorly understood. This study identifies Sonic hedgehog (Shh) signaling as a critical pathway in the transition from neurogenesis to oligodendrogenesis in dorsal forebrain progenitors during late embryonic development. The timing of this neuron-to-glia "switch" coincides with the arrival of migrating interneurons into the dorsal germinal zone, which we identify as a critical source of Shh ligand, which drives oligodendrogenesis. Our data provide evidence for a new model in which Shh signaling increases in the dorsal forebrain late in embryonic development to provide a temporally regulated mechanism that initiates the third wave of neocortical oligodendrogenesis.
Collapse
|
32
|
Farreny MA, Agius E, Bel-Vialar S, Escalas N, Khouri-Farah N, Soukkarieh C, Danesin C, Pituello F, Cochard P, Soula C. FGF signaling controls Shh-dependent oligodendroglial fate specification in the ventral spinal cord. Neural Dev 2018. [PMID: 29519242 PMCID: PMC5842613 DOI: 10.1186/s13064-018-0100-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background Most oligodendrocytes of the spinal cord originate from ventral progenitor cells of the pMN domain, characterized by expression of the transcription factor Olig2. A minority of oligodendrocytes is also recognized to emerge from dorsal progenitors during fetal development. The prevailing view is that generation of ventral oligodendrocytes depends on Sonic hedgehog (Shh) while dorsal oligodendrocytes develop under the influence of Fibroblast Growth Factors (FGFs). Results Using the well-established model of the chicken embryo, we show that ventral spinal progenitor cells activate FGF signaling at the onset of oligodendrocyte precursor cell (OPC) generation. Inhibition of FGF receptors at that time appears sufficient to prevent generation of ventral OPCs, highlighting that, in addition to Shh, FGF signaling is required also for generation of ventral OPCs. We further reveal an unsuspected interplay between Shh and FGF signaling by showing that FGFs serve dual essential functions in ventral OPC specification. FGFs are responsible for timely induction of a secondary Shh signaling center, the lateral floor plate, a crucial step to create the burst of Shh required for OPC specification. At the same time, FGFs prevent down-regulation of Olig2 in pMN progenitor cells as these cells receive higher threshold of the Shh signal. Finally, we bring arguments favoring a key role of newly differentiated neurons acting as providers of the FGF signal required to trigger OPC generation in the ventral spinal cord. Conclusion Altogether our data reveal that the FGF signaling pathway is activated and required for OPC commitment in the ventral spinal cord. More generally, our data may prove important in defining strategies to produce large populations of determined oligodendrocyte precursor cells from undetermined neural progenitors, including stem cells. In the long run, these new data could be useful in attempts to stimulate the oligodendrocyte fate in residing neural stem cells.
Collapse
Affiliation(s)
- Marie-Amélie Farreny
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Eric Agius
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Sophie Bel-Vialar
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Nathalie Escalas
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Nagham Khouri-Farah
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Chadi Soukkarieh
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Cathy Danesin
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Fabienne Pituello
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Philippe Cochard
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France
| | - Cathy Soula
- Centre de Biologie du Développement (CBD) CNRS/UPS, Centre de Biologie Intégrative (CBI), Université de Toulouse, F-31062, Toulouse, France.
| |
Collapse
|
33
|
Memi F, Zecevic N, Radonjić N. Multiple roles of Sonic Hedgehog in the developing human cortex are suggested by its widespread distribution. Brain Struct Funct 2018; 223:2361-2375. [PMID: 29492654 PMCID: PMC5968052 DOI: 10.1007/s00429-018-1621-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/25/2018] [Indexed: 01/01/2023]
Abstract
Sonic Hedgehog (Shh) plays an instrumental role in brain development, fine-tuning processes such as cell proliferation, patterning, and fate specification. Although, mutations in the SHH pathway in humans are associated with various neurodevelopmental disorders, ranging from holoprosencephaly to schizophrenia, its expression pattern in the developing human brain is not well established. We now determined the previously not reported wide expression of SHH in the human fetal cerebral cortex during most of the gestation period (10–40 gestational weeks). This spatiotemporal distribution puts Shh in a position to influence the fundamental processes involved in corticogenesis. SHH expression increased during development, shifting from progenitor cells in the proliferative zones to neurons, both glutamatergic and GABAergic, and astrocytes in upper cortical compartments. Importantly, the expression of its downstream effectors and complementary receptors revealed evolutionary differences in SHH-pathway gene expression between humans and rodents.
Collapse
Affiliation(s)
- Fani Memi
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA. .,Department of Cell and Developmental Biology, University College London, 21 University Street, London, WC1E 6DE, UK.
| | - Nada Zecevic
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Nevena Radonjić
- Department of Psychiatry, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
34
|
Chi L, Fan B, Feng D, Chen Z, Liu Z, Hui Y, Xu X, Ma L, Fang Y, Zhang Q, Jin G, Liu L, Guan F, Zhang X. The Dorsoventral Patterning of Human Forebrain Follows an Activation/Transformation Model. Cereb Cortex 2018; 27:2941-2954. [PMID: 27226442 DOI: 10.1093/cercor/bhw152] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The anteroposterior patterning of the central nervous system follows an activation/transformation model, which proposes that a prospective telencephalic fate will be activated by default during the neural induction stage, while this anterior fate could be transformed posteriorly according to caudalization morphogens. Although both extrinsic signals and intrinsic transcription factors have been implicated in dorsoventral (DV) specification of vertebrate telencephalon, the DV patterning model remains elusive. This is especially true in human considering its evolutionary trait and uniqueness of gene regulatory networks during neural induction. Here, we point to a model that human forebrain DV patterning also follows an activation/transformation paradigm. Human neuroectoderm (NE) will activate a forebrain dorsal fate automatically and this default anterior dorsal fate does not depend on Wnts activation or Pax6 expression. Forced expression of Pax6 in human NE hinders its ventralization even under sonic hedgehog (Shh) treatment, suggesting that the ventral fate is repressed by dorsal genes. Genetic manipulation of Nkx2.1, a key gene for forebrain ventral progenitors, shows that Nkx2.1 is neither necessary nor sufficient for Shh-driven ventralization. We thus propose that Shh represses dorsal genes of human NE and subsequently transforms the primitively activated dorsal fate ventrally in a repression release manner.
Collapse
Affiliation(s)
- Liankai Chi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences and.,School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China.,Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Beibei Fan
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Dandan Feng
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhenyu Chen
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Zhongliang Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Yi Hui
- Department of Anatomy and Neurobiology, The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Jiangsu 226001, China
| | - Xiangjie Xu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Lin Ma
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Yujiang Fang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Quanbin Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China
| | - Guohua Jin
- Department of Anatomy and Neurobiology, The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Jiangsu 226001, China
| | - Ling Liu
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China.,Advanced Institute of Translational Medicine
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Xiaoqing Zhang
- Shanghai Tenth People's Hospital, and Neuroregeneration Key Laboratory of Shanghai Universities, Tongji University School of Medicine, Shanghai 200092, China.,Advanced Institute of Translational Medicine.,The Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China
| |
Collapse
|
35
|
Abstract
The ZIC2 transcription factor is one of the most commonly mutated genes in Holoprosencephaly (HPE) probands. HPE is a severe congenital defect of forebrain development which occurs when the cerebral hemispheres fail to separate during the early stages of organogenesis and is typically associated with mispatterning of the embryonic midline. Recent study of genotype-phenotype correlations in HPE cases has defined distinctive features of ZIC2-associated HPE presentation and genetics, revealing that ZIC2 mutation does not produce the craniofacial abnormalities generally thought to characterise HPE but leads to a range of non-forebrain phenotypes. Furthermore, the studies confirm the extent of ZIC2 allelic heterogeneity and that pathogenic variants of ZIC2 are associated with both classic and middle interhemispheric variant (MIHV) HPE which arise from defective ventral and dorsal forebrain patterning, respectively. An allelic series of mouse mutants has helped to delineate the cellular and molecular mechanisms by which one gene leads to defects in these related but distinct embryological processes.
Collapse
Affiliation(s)
- Kristen S Barratt
- Early Mammalian Development Laboratory, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Ruth M Arkell
- Early Mammalian Development Laboratory, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
36
|
Adnani L, Han S, Li S, Mattar P, Schuurmans C. Mechanisms of Cortical Differentiation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:223-320. [DOI: 10.1016/bs.ircmb.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
37
|
Agirman G, Broix L, Nguyen L. Cerebral cortex development: an outside‐in perspective. FEBS Lett 2017; 591:3978-3992. [DOI: 10.1002/1873-3468.12924] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Gulistan Agirman
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| | - Loïc Broix
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| | - Laurent Nguyen
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| |
Collapse
|
38
|
Turrero García M, Harwell CC. Radial glia in the ventral telencephalon. FEBS Lett 2017; 591:3942-3959. [PMID: 28862741 DOI: 10.1002/1873-3468.12829] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/31/2022]
Abstract
The ventral telencephalon is the developmental origin of the basal ganglia and the source of neuronal and glial cells that integrate into developing circuits in other areas of the brain. Radial glia in the embryonic subpallium give rise to an enormous diversity of mature cell types, either directly or through other transit-amplifying progenitors. Here, we review current knowledge about these subpallial neural stem cells and their progeny, focusing on the period of neurogenesis. We describe their cell biological features and the extrinsic and intrinsic molecular codes that guide their fate specification in defined temporal and spatial sequences. We also discuss the role of clonal lineage in the organization and specification of mature neurons.
Collapse
Affiliation(s)
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Fedele S, Collo G, Behr K, Bischofberger J, Müller S, Kunath T, Christensen K, Gündner AL, Graf M, Jagasia R, Taylor V. Expansion of human midbrain floor plate progenitors from induced pluripotent stem cells increases dopaminergic neuron differentiation potential. Sci Rep 2017; 7:6036. [PMID: 28729666 PMCID: PMC5519680 DOI: 10.1038/s41598-017-05633-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/31/2017] [Indexed: 11/09/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are invaluable to study developmental processes and disease mechanisms particularly in the brain. hiPSCs can be differentiated into mature and functional dopaminergic (DA) neurons. Having robust protocols for the generation of differentiated DA neurons from pluripotent cells is a prerequisite for the use of hiPSCs to study disease mechanisms, for drug discovery, and eventually for cell replacement therapy. Here, we describe a protocol for generating and expanding large numbers of homogeneous midbrain floor plate progenitors (mFPPs) that retain efficient DA neurogenic potential over multiple passages and can be cryobanked. We demonstrate that expanded mFPPs have increased DA neuron potential and differentiate more efficiently and rapidly than progenitors generated by standard protocols. In addition, this novel method results in increased numbers of DA neurons that in vitro show characteristic electrophysiological properties of nigrostriatal DA neurons, produce high levels of dopamine, and integrate into host mice when grafted in vivo. Thus, we describe a robust method for producing human mesencephalic DA neurons from hiPSCs.
Collapse
Affiliation(s)
- Stefania Fedele
- Department of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058, Basel, Switzerland
| | - Ginetta Collo
- Department of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058, Basel, Switzerland.,Department of Molecular and Translational Medicine, Viale Europa 11, 25123, Brescia, Italy
| | - Katharina Behr
- Department of Biomedicine, University of Basel, Pestalozzistrasse 20, CH-4056, Basel, Switzerland
| | - Josef Bischofberger
- Department of Biomedicine, University of Basel, Pestalozzistrasse 20, CH-4056, Basel, Switzerland
| | - Stephan Müller
- Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Tilo Kunath
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, EH93JQ, Edinburgh, United Kingdom
| | - Klaus Christensen
- Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Anna Lisa Gündner
- Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Martin Graf
- Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Ravi Jagasia
- Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058, Basel, Switzerland.
| |
Collapse
|
40
|
Horiuchi M, Suzuki-Horiuchi Y, Akiyama T, Itoh A, Pleasure D, Carstens E, Itoh T. Differing intrinsic biological properties between forebrain and spinal oligodendroglial lineage cells. J Neurochem 2017; 142:378-391. [PMID: 28512742 DOI: 10.1111/jnc.14074] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/21/2022]
Abstract
Differentiation of oligodendroglial progenitor cells (OPCs) into myelinating oligodendrocytes is known to be regulated by the microenvironment where they differentiate. However, current research has not verified whether or not oligodendroglial lineage cells (OLCs) derived from different anatomical regions of the central nervous system (CNS) respond to microenvironmental cues in the same manner. Here, we isolated pure OPCs from rat neonatal forebrain (FB) and spinal cord (SC) and compared their phenotypes in the same in vitro conditions. We found that although FB and SC OLCs responded differently to the same external factors; they were distinct in proliferation response to mitogens, oligodendrocyte phenotype after differentiation, and cytotoxic responses to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate-type glutamate receptor-mediated excitotoxicity at immature stages of differentiation in a cell-intrinsic manner. Moreover, transcriptome analysis identified genes differentially expressed between these OPC populations, including those encoding transcription factors (TFs), cell surface molecules, and signaling molecules. Particularly, FB and SC OPCs retained the expression of FB- or SC-specific TFs, such as Foxg1 and Hoxc8, respectively, even after serial passaging in vitro. Given the essential role of these TFs in the regional identities of CNS cells along the rostrocaudal axis, our results suggest that CNS region-specific gene regulation by these TFs may cause cell-intrinsic differences in cellular responses between FB and SC OLCs to extracellular molecules. Further understanding of the regional differences among OPC populations will help to improve treatments for demyelination in different CNS regions and to facilitate the development of stem cell-derived OPCs for cell transplantation therapies for demyelination. Cover Image for this issue: doi. 10.1111/jnc.13809.
Collapse
Affiliation(s)
- Makoto Horiuchi
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yoko Suzuki-Horiuchi
- Department of Dermatology, Institute of Regenerative Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tasuku Akiyama
- Temple Itch Center, Department of Dermatology, Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Aki Itoh
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, California, USA.,Department of Neurology, School of Medicine, University of California, Sacramento, California, USA
| | - David Pleasure
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, California, USA.,Department of Neurology, School of Medicine, University of California, Sacramento, California, USA
| | - Earl Carstens
- Department of Neurobiology, Physiology & Behavior, University of California, Davis, California, USA
| | - Takayuki Itoh
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, California, USA.,Department of Neurology, School of Medicine, University of California, Sacramento, California, USA
| |
Collapse
|
41
|
Kadoshima T, Sakaguchi H, Eiraku M. Generation of Various Telencephalic Regions from Human Embryonic Stem Cells in Three-Dimensional Culture. Methods Mol Biol 2017; 1597:1-16. [PMID: 28361306 DOI: 10.1007/978-1-4939-6949-4_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In the developing embryo, telencephalon arises from the rostral portion of the neural tube. The telencephalon further subdivides into distinct brain regions along the dorsal-ventral (DV) axis by exogenous patterning signals. Here, we describe a protocol for in vitro generation of various telencephalic regions from human embryonic stem cells (ESCs). Dissociated human ESCs are reaggregated in a low-cell-adhesion 96-well plate and cultured as floating aggregates. Telencephalic neural progenitors are efficiently generated when ESC aggregates are cultured in serum-free medium containing TGFβ inhibitor and Wnt inhibitor. In long-term culture, the telencephalic neural progenitors acquire cortical identities and self-organize a stratified cortical structure as seen in human fetal cortex. By treatment with Shh signal, the telencephalic progenitors acquire ventral (subpallial) identities and generate lateral ganglionic eminence (LGE) and medial ganglionic eminence (MGE). In contrast, by treatment with Wnt and BMP signals, their regional identities shift to more dorsal side that generates choroid plexus and medial palllium (hippocampal primordium).
Collapse
Affiliation(s)
- Taisuke Kadoshima
- Cell Asymmetry team, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
- Asubio Pharma Co., Ltd., Kobe, Hyogo, Japan
| | - Hideya Sakaguchi
- In Vitro Histogenesis team, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Mototsugu Eiraku
- In Vitro Histogenesis team, RIKEN Center for Developmental Biology, Kobe, Hyogo, Japan
| |
Collapse
|
42
|
Ansen-Wilson LJ, Lipinski RJ. Gene-environment interactions in cortical interneuron development and dysfunction: A review of preclinical studies. Neurotoxicology 2017; 58:120-129. [PMID: 27932026 PMCID: PMC5328258 DOI: 10.1016/j.neuro.2016.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 12/03/2016] [Accepted: 12/03/2016] [Indexed: 12/26/2022]
Abstract
Cortical interneurons (cINs) are a diverse group of locally projecting neurons essential to the organization and regulation of neural networks. Though they comprise only ∼20% of neurons in the neocortex, their dynamic modulation of cortical activity is requisite for normal cognition and underlies multiple aspects of learning and memory. While displaying significant morphological, molecular, and electrophysiological variability, cINs collectively function to maintain the excitatory-inhibitory balance in the cortex by dampening hyperexcitability and synchronizing activity of projection neurons, primarily through use of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). Disruption of the excitatory-inhibitory balance is a common pathophysiological feature of multiple seizure and neuropsychiatric disorders, including epilepsy, schizophrenia, and autism. While most studies have focused on genetic disruption of cIN development in these conditions, emerging evidence indicates that cIN development is exquisitely sensitive to teratogenic disruption. Here, we review key aspects of cIN development, including specification, migration, and integration into neural circuits. Additionally, we examine the mechanisms by which prenatal exposure to common chemical and environmental agents disrupt these events in preclinical models. Understanding how genetic and environmental factors interact to disrupt cIN development and function has tremendous potential to advance prevention and treatment of prevalent seizure and neuropsychiatric illnesses.
Collapse
Affiliation(s)
- Lydia J Ansen-Wilson
- Department of Comparative Biosciences School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| | - Robert J Lipinski
- Department of Comparative Biosciences School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA; Molecular and Environmental Toxicology Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1010B McArdle Building, 1400 University Avenue, Madison, WI, 53706, USA.
| |
Collapse
|
43
|
Naruse M, Ishizaki Y, Ikenaka K, Tanaka A, Hitoshi S. Origin of oligodendrocytes in mammalian forebrains: a revised perspective. J Physiol Sci 2017; 67:63-70. [PMID: 27573166 PMCID: PMC5368213 DOI: 10.1007/s12576-016-0479-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) appear in the late embryonic brain, mature into oligodendrocytes (OLs), and form myelin in the postnatal brain. It has been proposed that early born OPCs derived from the ventral forebrain are eliminated postnatally and late-born OLs predominate in the adult mouse cortex. However, the temporal and regional niche for cortical OL generation, which persists throughout life in adult mammals, remains to be determined. Our recent study provides new insight into self-renewing and multipotent neural stem cells (NSCs). Our results, together with previous studies, suggest that NSCs at the dorsoventral boundary are uniquely specialized to produce myelin-forming OLs in the cortex during a restricted temporal window. These findings may help identify transcription factors or gene expression patterns which confer neural precursors with the characteristic ability of dorsoventral boundary NSCs to differentiate into OLs, and facilitate the development of new strategies for regenerative medicine of the damaged brain.
Collapse
Affiliation(s)
- Masae Naruse
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| | - Aoi Tanaka
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Seiji Hitoshi
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan.
| |
Collapse
|
44
|
Zhou X, Pace J, Filichia E, Lv T, Davis B, Hoffer B, Selman W, Luo Y. Effect of the sonic hedgehog receptor smoothened on the survival and function of dopaminergic neurons. Exp Neurol 2016; 283:235-45. [PMID: 27317298 PMCID: PMC5479305 DOI: 10.1016/j.expneurol.2016.06.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/24/2016] [Accepted: 06/12/2016] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To determine the influence of the sonic hedgehog (shh) pathway and its receptor smoothened (smo), on the survival and functionality of dopaminergic (DA) neurons. BACKGROUND During early development, shh induces the differentiation of DA neurons. However, it is unknown whether shh signaling is required in the maturation or maintenance of DA neurons during later development and adulthood due to the lethality of traditional shh knockout models. METHODS We utilized the cre-loxP system to achieve late developmental stage and cell type-specific deletion of the shh receptor, smo, in DA neurons by crossing DATcre (dopamine transporter) mice with Smo(loxP/loxP) mice. We assessed for differences between knockout (ko) and wildtype (wt) mice using combined histochemistry, gene expression analysis, and behavioral evaluation. Number and size of DA neurons in ventral midbrain and the DA neural terminal density in striatum were measured using unbiased stereological quantification. The survival of DA neurons under neurotoxin challenge was examined in the unilateral 6-hydroxydopamine (6-OHDA) Parkinson's disease animal model and the more subtle function under challenge of the dopaminergic system was examined by methamphetamine single- and repeated challenge in wt and ko mice. RESULTS Tyrosine hydroxylase (TH) positive neuronal counts and neuronal size in substantia nigra (SN) and ventral tegmental area (VTA) showed no difference between wt and DAT-Smo ko mice in young (5months) or aged (22months) mice. There was also no difference in the striatal DA projections between wt and ko mice in both age groups. In unilateral striatal 6-OHDA lesions modeling Parkinson's disease, using stereotaxic injection of 6-OHDA intrastriatally led to loss of dopaminergic neurons in SN and diminished TH positive projections in striatum. However, there was no differences in survival of DA neurons between wt and ko mice. DAT-Smo ko mice demonstrated hyperactivity compared to wt mice at 5months, but showed no difference in activity at 22months. When injected with a one-time bolus of methamphetamine (METH), despite the higher basal locomotion activity, DAT-Smo ko mice showed a diminished response to a single METH challenge. In METH sensitization testing, ko mice showed decreased sensitization compared to wt mice without evidence of a delayed shift in dynamics of sensitization. Gene expression analysis showed decreased gene expression of smo, Gli 1 (known target gene of smo) and BDNF (brain-derived neurotrophic factor) in the SN. Gene expression was not altered in striatum for the genes examined in this study including dopamine receptor genes, neurotropic genes such as Glial cell line-derived neurotrophic factor (GDNF), and bone morphogenetic protein 7 (BMP7). CONCLUSION Our study showed the smo receptor function is not required for the maturation and survival of DA neurons during late development, aging or under stress challenge. However, smo function has an influence on behavior in young adult mice and in responses of mice to a drug that modulates DA neurochemistry through regulation of gene expression in DA neurons. Since young adult DAT-smo ko mice show hyperactivity and altered response to a psychostimulant drug (METH), this may indicate the involvement of the shh pathway in the development of functional changes that manifest as alterations in DA pathway dynamics.
Collapse
Affiliation(s)
- Xiaofei Zhou
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Jonathan Pace
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Emily Filichia
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Tao Lv
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Brandon Davis
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Barry Hoffer
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Warren Selman
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA
| | - Yu Luo
- Department of Neurological Surgery, University Hospitals Case Medical Center, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
45
|
Ahn S, Kim TG, Kim KS, Chung S. Differentiation of human pluripotent stem cells into Medial Ganglionic Eminence vs. Caudal Ganglionic Eminence cells. Methods 2016; 101:103-12. [PMID: 26364591 PMCID: PMC4786472 DOI: 10.1016/j.ymeth.2015.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/29/2015] [Accepted: 09/09/2015] [Indexed: 12/18/2022] Open
Abstract
Human pluripotent stem cells (PSCs) represent an opportunity to study human development in vitro, to model diseases in a dish, to screen drugs as well as to provide an unlimited and ethically unimpeded source of therapeutic cells. Cortical GABAergic interneurons, which are generated from Medial Ganglionic Eminence (MGE) cells and Caudal Ganglionic Eminence (CGE) cells during embryonic development, regulate cortical neural networks by providing inhibitory inputs. Their malfunction, resulting in failure to intricately regulate neural circuit balance, has been implicated in brain diseases, such as schizophrenia, autism and epilepsy. In this study, using combinatorial and temporal modulation of developmentally relevant dorsoventral and rostrocaudal signaling pathways, we efficiently generated MGE cells vs. CGE cells from human PSCs, which predominantly generate Parvalbumin-expressing or Somatostatin-expressing interneurons vs. Calretinin-expressing interneurons, respectively. Efficient generation of specific differentiated progenies of hPSCs as shown in this study will be a pivotal step to realize the full potential of hPSCs for regenerative medicine, developmental studies, disease modeling, bioassay, and drug screening.
Collapse
Affiliation(s)
- Sandra Ahn
- Translational Stem Cell Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Tae-Gon Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA
| | - Sangmi Chung
- Translational Stem Cell Neurobiology Laboratory, Department of Psychiatry and Program in Neuroscience, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA; Harvard Stem Cell Institute, McLean Hospital/Harvard Medical School, Belmont, MA 02478, USA.
| |
Collapse
|
46
|
Delgado RN, Lu C, Lim DA. Maintenance of neural stem cell regional identity in culture. NEUROGENESIS 2016; 3:e1187321. [PMID: 27606338 DOI: 10.1080/23262133.2016.1187321] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/25/2016] [Accepted: 05/03/2016] [Indexed: 01/01/2023]
Abstract
Neural stem cells (NSCs) are distributed throughout the ventricular-subventricular zone (V-SVZ) in the adult mouse brain. NSCs located in spatially distinct regions of the V-SVZ generate different types of olfactory bulb (OB) neurons, and the regional expression of specific transcription factors correlates with these differences in NSC developmental potential. In a recent article, we show that Nkx2.1-expressing embryonic precursors give rise to NKX2.1+ NSCs located in the ventral V-SVZ of adult mice. Here we characterize a V-SVZ monolayer culture system that retains regional gene expression and neurogenic potential of NSCs from the dorsal and ventral V-SVZ. In particular, we find that Nkx2.1-lineage V-SVZ NSCs maintain Nkx2.1 expression through serial passage and can generate new neurons in vitro. Thus, V-SVZ NSCs retain key aspects of their in vivo regional identity in culture, providing new experimental opportunities for understanding how such developmental patterns are established and maintained during development.
Collapse
Affiliation(s)
- Ryan N Delgado
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA, USA; Biomedical Sciences Program, University of California at San Francisco, San Francisco, CA, USA; Medical Scientist Training Program, University of California at San Francisco, San Francisco, CA, USA
| | - Changqing Lu
- Department of Anatomy, Premedical and Forensic School, Sichuan University, Chengdu, China; Department of Forensic Toxicological Analysis, Premedical and Forensic School, Sichuan University, Chengdu, China
| | - Daniel A Lim
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA, USA; San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| |
Collapse
|
47
|
Kawaguchi D, Sahara S, Zembrzycki A, O'Leary DDM. Generation and analysis of an improved Foxg1-IRES-Cre driver mouse line. Dev Biol 2016; 412:139-147. [PMID: 26896590 DOI: 10.1016/j.ydbio.2016.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 11/28/2022]
Abstract
Foxg1 expression is highly restricted to the telencephalon and other head structures in the early embryo. This expression pattern has been exploited to generate conditional knockout mice, based on a widely used Foxg1-Cre knock-in line (Foxg1(tm1(cre)Skm)), in which the Foxg1 coding region was replaced by the Cre gene. The utility of this line, however, is severely hampered for two reasons: (1) Foxg1-Cre mice display ectopic and unpredictable Cre activity, and (2) Foxg1 haploinsufficiency can produce neurodevelopmental phenotypes. To overcome these issues, we have generated a new Foxg1-IRES-Cre knock-in mouse line, in which an IRES-Cre cassette was inserted in the 3'UTR of Foxg1 locus, thus preserving the endogenous Foxg1 coding region and un-translated gene regulatory sequences in the 3'UTR, including recently discovered microRNA target sites. We further demonstrate that the new Foxg1-IRES-Cre line displays consistent Cre activity patterns that recapitulated the endogenous Foxg1 expression at embryonic and postnatal stages without causing defects in cortical development. We conclude that the new Foxg1-IRES-Cre mouse line is a unique and advanced tool for studying genes involved in the development of the telencephalon and other Foxg1-expressing regions starting from early embryonic stages.
Collapse
Affiliation(s)
- Daichi Kawaguchi
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | - Setsuko Sahara
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Andreas Zembrzycki
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Dennis D M O'Leary
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
48
|
Tyson JA, Goldberg EM, Maroof AM, Xu Q, Petros TJ, Anderson SA. Duration of culture and sonic hedgehog signaling differentially specify PV versus SST cortical interneuron fates from embryonic stem cells. Development 2016; 142:1267-78. [PMID: 25804737 DOI: 10.1242/dev.111526] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Medial ganglionic eminence (MGE)-derived GABAergic cortical interneurons (cINs) consist of multiple subtypes that are involved in many cortical functions. They also have a remarkable capacity to migrate, survive and integrate into cortical circuitry after transplantation into postnatal cortex. These features have engendered considerable interest in generating distinct subgroups of interneurons from pluripotent stem cells (PSCs) for the study of interneuron fate and function, and for the development of cell-based therapies. Although advances have been made, the capacity to generate highly enriched pools of subgroup fate-committed interneuron progenitors from PSCs has remained elusive. Previous studies have suggested that the two main MGE-derived interneuron subgroups--those expressing somatostatin (SST) and those expressing parvalbumin (PV)--are specified in the MGE from Nkx2.1-expressing progenitors at higher or lower levels of sonic hedgehog (Shh) signaling, respectively. To further explore the role of Shh and other factors in cIN fate determination, we generated a reporter line such that Nkx2.1-expressing progenitors express mCherry and postmitotic Lhx6-expressing MGE-derived interneurons express GFP. Manipulations of Shh exposure and time in culture influenced the subgroup fates of ESC-derived interneurons. Exposure to higher Shh levels, and collecting GFP-expressing precursors at 12 days in culture, resulted in the strongest enrichment for SST interneurons over those expressing PV, whereas the strongest enrichment for PV interneurons was produced by lower Shh and by collecting mCherry-expressing cells after 17 days in culture. These findings confirm that fate determination of cIN subgroups is crucially influenced by Shh signaling, and provide a system for the further study of interneuron fate and function.
Collapse
Affiliation(s)
- Jennifer A Tyson
- Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine ARC 517, Philadelphia, PA 19104-5127, USA Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Ethan M Goldberg
- Division of Neurology, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA 19083, USA Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19083, USA
| | - Asif M Maroof
- Harvard University Department of Stem Cell and Regenerative Biology, Cambridge, MA 02138, USA
| | - Qing Xu
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Timothy J Petros
- Department of Neuroscience, NYU Langone Medical Center, New York, NY 10016, USA
| | - Stewart A Anderson
- Department of Psychiatry, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine ARC 517, Philadelphia, PA 19104-5127, USA
| |
Collapse
|
49
|
Choe Y, Pleasure SJ, Mira H. Control of Adult Neurogenesis by Short-Range Morphogenic-Signaling Molecules. Cold Spring Harb Perspect Biol 2015; 8:a018887. [PMID: 26637286 DOI: 10.1101/cshperspect.a018887] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Adult neurogenesis is dynamically regulated by a tangled web of local signals emanating from the neural stem cell (NSC) microenvironment. Both soluble and membrane-bound niche factors have been identified as determinants of adult neurogenesis, including morphogens. Here, we review our current understanding of the role and mechanisms of short-range morphogen ligands from the Wnt, Notch, Sonic hedgehog, and bone morphogenetic protein (BMP) families in the regulation of adult neurogenesis. These morphogens are ideally suited to fine-tune stem-cell behavior, progenitor expansion, and differentiation, thereby influencing all stages of the neurogenesis process. We discuss cross talk between their signaling pathways and highlight findings of embryonic development that provide a relevant context for understanding neurogenesis in the adult brain. We also review emerging examples showing that the web of morphogens is in fact tightly linked to the regulation of neurogenesis by diverse physiologic processes.
Collapse
Affiliation(s)
- Youngshik Choe
- Department of Neurology, Programs in Neuroscience, Developmental and Stem Cell Biology, UCSF Institute for Regeneration Medicine, San Francisco, California 94158
| | - Samuel J Pleasure
- Department of Neurology, Programs in Neuroscience, Developmental and Stem Cell Biology, UCSF Institute for Regeneration Medicine, San Francisco, California 94158
| | - Helena Mira
- Chronic Disease Programme, UFIEC, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| |
Collapse
|
50
|
Delgado RN, Lim DA. Embryonic Nkx2.1-expressing neural precursor cells contribute to the regional heterogeneity of adult V-SVZ neural stem cells. Dev Biol 2015; 407:265-74. [PMID: 26387477 DOI: 10.1016/j.ydbio.2015.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 08/02/2015] [Accepted: 09/15/2015] [Indexed: 01/10/2023]
Abstract
The adult ventricular-subventricular zone (V-SVZ) of the lateral ventricle produces several subtypes of olfactory bulb (OB) interneurons throughout life. Neural stem cells (NSCs) within this zone are heterogeneous, with NSCs located in different regions of the lateral ventricle wall generating distinct OB interneuron subtypes. The regional expression of specific transcription factors appears to correspond to such geographical differences in the developmental potential of V-SVZ NSCs. However, the transcriptional definition and developmental origin of V-SVZ NSC regional identity are not well understood. In this study, we found that a population of NSCs in the ventral region of the V-SVZ expresses the transcription factor Nkx2.1 and is derived from Nkx2.1-expressing (Nkx2.1+) embryonic precursors. To follow the fate of Nkx2.1+ cells and their progeny in vivo, we used mice with an Nkx2.1-CreER "knock-in" allele. Nkx2.1+ V-SVZ NSCs labeled in adult mice generated interneurons for the deep granule cell layer of the OB. Embryonic brain Nkx2.1+ precursors labeled at embryonic day 12.5 gave rise to Nkx2.1+ NSCs of the ventral V-SVZ in postnatal and adult mice. Thus, embryonic Nkx2.1+ neural precursors give rise to a population of Nkx2.1+ NSCs in the ventral V-SVZ where they contribute to the regional heterogeneity of V-SVZ NSCs.
Collapse
Affiliation(s)
- Ryan N Delgado
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Program, University of California at San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, University of California at San Francisco, San Francisco, CA 94143, USA
| | - Daniel A Lim
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA; San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA..
| |
Collapse
|