1
|
Jinnou H, Rosko LM, Yamashita S, Henmi S, Prasad J, Lam VK, Agaronyan A, Tu TW, Imamura Y, Kuboyama K, Sawamoto K, Hashimoto-Torii K, Ishibashi N, Gallo V. Outer radial glia promotes white matter regeneration after neonatal brain injury. Cell Rep Med 2025; 6:101986. [PMID: 40023165 PMCID: PMC11970391 DOI: 10.1016/j.xcrm.2025.101986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 11/27/2024] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
The developing gyrencephalic brain contains a large population of neural stem cells in the ventricular zone and outer subventricular zone (OSVZ), the latter populated by outer radial glia (oRG). The role of oRG during postnatal development is not well understood. We show that oRG cells increase proliferative capacity and contribute to oligodendrocyte precursor cell (OPC) production following brain injury in human infants and neonatal piglets, whose brains resemble the human brain in structure and development. RNA sequencing revealed oRG-specific transcriptional responses to injury in piglets and showed that the activating transcription factor 5 (ATF5) pathway positively regulates oRG proliferation. Intranasal activation of ATF5 using salubrinal enhanced OSVZ-derived oligodendrogenesis in the injured periventricular white matter and improved functional recovery. These results reveal a key role for postnatal oRG in brain injury recovery and identify ATF5 as a potential therapeutic target for treating white matter injury in infants.
Collapse
Affiliation(s)
- Hideo Jinnou
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA; Department of Pediatrics and Neonatology, Nagoya City University, Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Lauren M Rosko
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Satoshi Yamashita
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Soichiro Henmi
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Jaya Prasad
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Van K Lam
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Artur Agaronyan
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC 20059, USA
| | - Tsang-Wei Tu
- Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC 20059, USA
| | - Yuka Imamura
- Departments of Pharmacology and Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Kazuya Kuboyama
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Kazue Hashimoto-Torii
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA; Department of Pediatrics, Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA
| | - Nobuyuki Ishibashi
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA; Department of Pediatrics, Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA; Department of Pediatrics, Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA; Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
2
|
Namba T, Huttner WB. What Makes Us Human: Insights from the Evolution and Development of the Human Neocortex. Annu Rev Cell Dev Biol 2024; 40:427-452. [PMID: 39356810 DOI: 10.1146/annurev-cellbio-112122-032521] [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] [Indexed: 10/04/2024]
Abstract
"What makes us human?" is a central question of many research fields, notably anthropology. In this review, we focus on the development of the human neocortex, the part of the brain with a key role in cognition, to gain neurobiological insight toward answering this question. We first discuss cortical stem and progenitor cells and human-specific genes that affect their behavior. We thus aim to understand the molecular foundation of the expansion of the neocortex that occurred in the course of human evolution, as this expansion is generally thought to provide a basis for our unique cognitive abilities. We then review the emerging evidence pointing to differences in the development of the neocortex between present-day humans and Neanderthals, our closest relatives. Finally, we discuss human-specific genes that have been implicated in neuronal circuitry and offer a perspective for future studies addressing the question of what makes us human.
Collapse
Affiliation(s)
- Takashi Namba
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany;
| |
Collapse
|
3
|
Xing L, Huttner WB, Namba T. Role of cell metabolism in the pathophysiology of brain size-associated neurodevelopmental disorders. Neurobiol Dis 2024; 199:106607. [PMID: 39029564 DOI: 10.1016/j.nbd.2024.106607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/13/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024] Open
Abstract
Cell metabolism is a key regulator of human neocortex development and evolution. Several lines of evidence indicate that alterations in neural stem/progenitor cell (NPC) metabolism lead to abnormal brain development, particularly brain size-associated neurodevelopmental disorders, such as microcephaly. Abnormal NPC metabolism causes impaired cell proliferation and thus insufficient expansion of NPCs for neurogenesis. Therefore, the production of neurons, which is a major determinant of brain size, is decreased and the size of the brain, especially the size of the neocortex, is significantly reduced. This review discusses recent progress understanding NPC metabolism, focusing in particular on glucose metabolism, fatty acid metabolism and amino acid metabolism (e.g., glutaminolysis and serine metabolism). We provide an overview of the contributions of these metabolic pathways to brain development and evolution, as well as to the etiology of neurodevelopmental disorders. Furthermore, we discuss the advantages and disadvantages of various experimental models to study cell metabolism in the developing brain.
Collapse
Affiliation(s)
- Lei Xing
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada.
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland; Department of Developmental Biology, Fujita Health University School of Medicine, Toyoake, Japan; International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Japan.
| |
Collapse
|
4
|
Vivi E, Di Benedetto B. Brain stars take the lead during critical periods of early postnatal brain development: relevance of astrocytes in health and mental disorders. Mol Psychiatry 2024; 29:2821-2833. [PMID: 38553540 PMCID: PMC11420093 DOI: 10.1038/s41380-024-02534-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 09/25/2024]
Abstract
In the brain, astrocytes regulate shape and functions of the synaptic and vascular compartments through a variety of released factors and membrane-bound proteins. An imbalanced astrocyte activity can therefore have drastic negative impacts on brain development, leading to the onset of severe pathologies. Clinical and pre-clinical studies show alterations in astrocyte cell number, morphology, molecular makeup and astrocyte-dependent processes in different affected brain regions in neurodevelopmental (ND) and neuropsychiatric (NP) disorders. Astrocytes proliferate, differentiate and mature during the critical period of early postnatal brain development, a time window of elevated glia-dependent regulation of a proper balance between synapse formation/elimination, which is pivotal in refining synaptic connectivity. Therefore, any intrinsic and/or extrinsic factors altering these processes during the critical period may result in an aberrant synaptic remodeling and onset of mental disorders. The peculiar bridging position of astrocytes between synaptic and vascular compartments further allows them to "compute" the brain state and consequently secrete factors in the bloodstream, which may serve as diagnostic biomarkers of distinct healthy or disease conditions. Here, we collect recent advancements regarding astrogenesis and astrocyte-mediated regulation of neuronal network remodeling during early postnatal critical periods of brain development, focusing on synapse elimination. We then propose alternative hypotheses for an involvement of aberrancies in these processes in the onset of ND and NP disorders. In light of the well-known differential prevalence of certain brain disorders between males and females, we also discuss putative sex-dependent influences on these neurodevelopmental events. From a translational perspective, understanding age- and sex-dependent astrocyte-specific molecular and functional changes may help to identify biomarkers of distinct cellular (dys)functions in health and disease, favouring the development of diagnostic tools or the selection of tailored treatment options for male/female patients.
Collapse
Affiliation(s)
- Eugenia Vivi
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany
| | - Barbara Di Benedetto
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053, Regensburg, Germany.
- Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
5
|
Gkini V, Gómez-Lozano I, Heikinheimo O, Namba T. Dynamic changes in mitochondrial localization in human neocortical basal radial glial cells during cell cycle. J Comp Neurol 2024; 532:e25630. [PMID: 38852043 DOI: 10.1002/cne.25630] [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: 12/29/2023] [Revised: 03/25/2024] [Accepted: 05/09/2024] [Indexed: 06/10/2024]
Abstract
Mitochondria play critical roles in neural stem/progenitor cell proliferation and fate decisions. The subcellular localization of mitochondria in neural stem/progenitor cells during mitosis potentially influences the distribution of mitochondria to the daughter cells and thus their fates. Therefore, understanding the spatial dynamics of mitochondria provides important knowledge about brain development. In this study, we analyzed the subcellular localization of mitochondria in the fetal human neocortex with a particular focus on the basal radial glial cells (bRGCs), a neural stem/progenitor cell subtype attributed to the evolutionary expansion of the human neocortex. During interphase, bRGCs exhibit a polarized localization of mitochondria that is localized at the base of the process or the proximal part of the process. Thereafter, mitochondria in bRGCs at metaphase show unpolarized distribution in which the mitochondria are randomly localized in the cytoplasm. During anaphase and telophase, mitochondria are still localized evenly, but mainly in the periphery of the cytoplasm. Mitochondria start to accumulate at the cleavage furrow during cytokinesis. These results suggest that the mitochondrial localization in bRGCs is tightly regulated during the cell cycle, which may ensure the proper distribution of mitochondria to the daughter cells and, thus in turn, influence their fates.
Collapse
Affiliation(s)
- Vasiliki Gkini
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Inés Gómez-Lozano
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
6
|
Firth W, Pye KR, Weightman Potter PG. Astrocytes at the intersection of ageing, obesity, and neurodegeneration. Clin Sci (Lond) 2024; 138:515-536. [PMID: 38652065 DOI: 10.1042/cs20230148] [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: 11/17/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Once considered passive cells of the central nervous system (CNS), glia are now known to actively maintain the CNS parenchyma; in recent years, the evidence for glial functions in CNS physiology and pathophysiology has only grown. Astrocytes, a heterogeneous group of glial cells, play key roles in regulating the metabolic and inflammatory landscape of the CNS and have emerged as potential therapeutic targets for a variety of disorders. This review will outline astrocyte functions in the CNS in healthy ageing, obesity, and neurodegeneration, with a focus on the inflammatory responses and mitochondrial function, and will address therapeutic outlooks.
Collapse
Affiliation(s)
- Wyn Firth
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, U.K
| | - Katherine R Pye
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| | - Paul G Weightman Potter
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Faculty of Health and Life Sciences, University of Exeter, Exeter, U.K
| |
Collapse
|
7
|
Xing L, Gkini V, Nieminen AI, Zhou HC, Aquilino M, Naumann R, Reppe K, Tanaka K, Carmeliet P, Heikinheimo O, Pääbo S, Huttner WB, Namba T. Functional synergy of a human-specific and an ape-specific metabolic regulator in human neocortex development. Nat Commun 2024; 15:3468. [PMID: 38658571 PMCID: PMC11043075 DOI: 10.1038/s41467-024-47437-8] [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: 11/03/2023] [Accepted: 04/02/2024] [Indexed: 04/26/2024] Open
Abstract
Metabolism has recently emerged as a major target of genes implicated in the evolutionary expansion of human neocortex. One such gene is the human-specific gene ARHGAP11B. During human neocortex development, ARHGAP11B increases the abundance of basal radial glia, key progenitors for neocortex expansion, by stimulating glutaminolysis (glutamine-to-glutamate-to-alpha-ketoglutarate) in mitochondria. Here we show that the ape-specific protein GLUD2 (glutamate dehydrogenase 2), which also operates in mitochondria and converts glutamate-to-αKG, enhances ARHGAP11B's ability to increase basal radial glia abundance. ARHGAP11B + GLUD2 double-transgenic bRG show increased production of aspartate, a metabolite essential for cell proliferation, from glutamate via alpha-ketoglutarate and the TCA cycle. Hence, during human evolution, a human-specific gene exploited the existence of another gene that emerged during ape evolution, to increase, via concerted changes in metabolism, progenitor abundance and neocortex size.
Collapse
Affiliation(s)
- Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
- Department of Biological Sciences, University of Manitoba, Winnipeg, MB, Canada.
| | - Vasiliki Gkini
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Anni I Nieminen
- FIMM Metabolomics Unit, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Hui-Chao Zhou
- Center for Cancer Biology (CCB), VIB-KU Leuven, B-3000, Leuven, Belgium
| | - Matilde Aquilino
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Katrin Reppe
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, B-3000, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, B-3000, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Human Evolutionary Genomics Unit, Okinawa Institute of Science and Technology, Okinawa, Onna-son, Japan
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
8
|
Garone C, De Giorgio F, Carli S. Mitochondrial metabolism in neural stem cells and implications for neurodevelopmental and neurodegenerative diseases. J Transl Med 2024; 22:238. [PMID: 38438847 PMCID: PMC10910780 DOI: 10.1186/s12967-024-05041-w] [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: 12/29/2023] [Accepted: 02/25/2024] [Indexed: 03/06/2024] Open
Abstract
Mitochondria are cytoplasmic organelles having a fundamental role in the regulation of neural stem cell (NSC) fate during neural development and maintenance.During embryonic and adult neurogenesis, NSCs undergo a metabolic switch from glycolytic to oxidative phosphorylation with a rise in mitochondrial DNA (mtDNA) content, changes in mitochondria shape and size, and a physiological augmentation of mitochondrial reactive oxygen species which together drive NSCs to proliferate and differentiate. Genetic and epigenetic modifications of proteins involved in cellular differentiation (Mechanistic Target of Rapamycin), proliferation (Wingless-type), and hypoxia (Mitogen-activated protein kinase)-and all connected by the common key regulatory factor Hypoxia Inducible Factor-1A-are deemed to be responsible for the metabolic shift and, consequently, NSC fate in physiological and pathological conditions.Both primary mitochondrial dysfunction due to mutations in nuclear DNA or mtDNA or secondary mitochondrial dysfunction in oxidative phosphorylation (OXPHOS) metabolism, mitochondrial dynamics, and organelle interplay pathways can contribute to the development of neurodevelopmental or progressive neurodegenerative disorders.This review analyses the physiology and pathology of neural development starting from the available in vitro and in vivo models and highlights the current knowledge concerning key mitochondrial pathways involved in this process.
Collapse
Affiliation(s)
- C Garone
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy.
- IRCCS Istituto Delle Scienze Neurologiche di Bologna, UO Neuropsichiatria Dell'età Pediatrica, Bologna, Italy.
| | - F De Giorgio
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - S Carli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
9
|
Luppi AI, Girn M, Rosas FE, Timmermann C, Roseman L, Erritzoe D, Nutt DJ, Stamatakis EA, Spreng RN, Xing L, Huttner WB, Carhart-Harris RL. A role for the serotonin 2A receptor in the expansion and functioning of human transmodal cortex. Brain 2024; 147:56-80. [PMID: 37703310 DOI: 10.1093/brain/awad311] [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: 04/13/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/15/2023] Open
Abstract
Integrating independent but converging lines of research on brain function and neurodevelopment across scales, this article proposes that serotonin 2A receptor (5-HT2AR) signalling is an evolutionary and developmental driver and potent modulator of the macroscale functional organization of the human cerebral cortex. A wealth of evidence indicates that the anatomical and functional organization of the cortex follows a unimodal-to-transmodal gradient. Situated at the apex of this processing hierarchy-where it plays a central role in the integrative processes underpinning complex, human-defining cognition-the transmodal cortex has disproportionately expanded across human development and evolution. Notably, the adult human transmodal cortex is especially rich in 5-HT2AR expression and recent evidence suggests that, during early brain development, 5-HT2AR signalling on neural progenitor cells stimulates their proliferation-a critical process for evolutionarily-relevant cortical expansion. Drawing on multimodal neuroimaging and cross-species investigations, we argue that, by contributing to the expansion of the human cortex and being prevalent at the apex of its hierarchy in the adult brain, 5-HT2AR signalling plays a major role in both human cortical expansion and functioning. Owing to its unique excitatory and downstream cellular effects, neuronal 5-HT2AR agonism promotes neuroplasticity, learning and cognitive and psychological flexibility in a context-(hyper)sensitive manner with therapeutic potential. Overall, we delineate a dual role of 5-HT2ARs in enabling both the expansion and modulation of the human transmodal cortex.
Collapse
Affiliation(s)
- Andrea I Luppi
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
- Leverhulme Centre for the Future of Intelligence, University of Cambridge, Cambridge, CB2 1SB, UK
- The Alan Turing Institute, London, NW1 2DB, UK
| | - Manesh Girn
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
| | - Fernando E Rosas
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
- Data Science Institute, Imperial College London, London, SW7 2AZ, UK
- Centre for Complexity Science, Imperial College London, London, SW7 2AZ, UK
| | - Christopher Timmermann
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Leor Roseman
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David Erritzoe
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - David J Nutt
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Emmanuel A Stamatakis
- Department of Clinical Neurosciences and Division of Anaesthesia, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - R Nathan Spreng
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, H3A 2B4, Canada
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, 01307, Germany
| | - Robin L Carhart-Harris
- Psychedelics Division-Neuroscape, Department of Neurology, University of California SanFrancisco, San Francisco, CA 94158, USA
- Centre for Psychedelic Research, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
10
|
Akula SK, Exposito-Alonso D, Walsh CA. Shaping the brain: The emergence of cortical structure and folding. Dev Cell 2023; 58:2836-2849. [PMID: 38113850 PMCID: PMC10793202 DOI: 10.1016/j.devcel.2023.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/08/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The cerebral cortex-the brain's covering and largest region-has increased in size and complexity in humans and supports higher cognitive functions such as language and abstract thinking. There is a growing understanding of the human cerebral cortex, including the diversity and number of cell types that it contains, as well as of the developmental mechanisms that shape cortical structure and organization. In this review, we discuss recent progress in our understanding of molecular and cellular processes, as well as mechanical forces, that regulate the folding of the cerebral cortex. Advances in human genetics, coupled with experimental modeling in gyrencephalic species, have provided insights into the central role of cortical progenitors in the gyrification and evolutionary expansion of the cerebral cortex. These studies are essential for understanding the emergence of structural and functional organization during cortical development and the pathogenesis of neurodevelopmental disorders associated with cortical malformations.
Collapse
Affiliation(s)
- Shyam K Akula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - David Exposito-Alonso
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA.
| |
Collapse
|
11
|
An B, Ando A, Akuta H, Morishita F, Imamura T. Human-biased TMEM25 expression promotes expansion of neural progenitor cells to alter cortical structure in the developing brain. FEBS Lett 2023; 597:2611-2625. [PMID: 37846797 DOI: 10.1002/1873-3468.14756] [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: 05/09/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/18/2023]
Abstract
Cortical expansion has occurred during human brain evolution. By comparing human and mouse RNA-seq datasets, we found that transmembrane protein 25 (TMEM25) was much more highly expressed in human neural progenitors (NPCs). Overexpression of either human TMEM25 or mouse Tmem25 similarly promoted mouse NPC proliferation in vitro. Mimicking human-type expression of TMEM25 in mouse ventricular cortical progenitors accelerated proliferation of basal radial glia (bRG) and increased the number of upper-layer neurons in vivo. By contrast, RNA-seq analysis, and pharmacological assays showed that knockdown of TMEM25 in cultured human NPCs compromised the effects of extracellular signals, leading to cell cycle inhibition via Akt repression. Thus, TMEM25 can receive extracellular signals to expand bRG in human cortical development.
Collapse
Affiliation(s)
- Boyang An
- Laboratory of Molecular and Cellular Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Japan
| | - Akari Ando
- Laboratory of Molecular and Cellular Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Japan
| | - Hiroto Akuta
- Laboratory of Molecular and Cellular Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Japan
| | - Fumihiro Morishita
- Laboratory of Molecular and Cellular Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Japan
| | - Takuya Imamura
- Laboratory of Molecular and Cellular Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Japan
| |
Collapse
|
12
|
Weber AI, Parthasarathy S, Borisova E, Epifanova E, Preußner M, Rusanova A, Ambrozkiewicz MC, Bessa P, Newman A, Müller L, Schaal H, Heyd F, Tarabykin V. Srsf1 and Elavl1 act antagonistically on neuronal fate choice in the developing neocortex by controlling TrkC receptor isoform expression. Nucleic Acids Res 2023; 51:10218-10237. [PMID: 37697438 PMCID: PMC10602877 DOI: 10.1093/nar/gkad703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 07/24/2023] [Accepted: 08/15/2023] [Indexed: 09/13/2023] Open
Abstract
The seat of higher-order cognitive abilities in mammals, the neocortex, is a complex structure, organized in several layers. The different subtypes of principal neurons are distributed in precise ratios and at specific positions in these layers and are generated by the same neural progenitor cells (NPCs), steered by a spatially and temporally specified combination of molecular cues that are incompletely understood. Recently, we discovered that an alternatively spliced isoform of the TrkC receptor lacking the kinase domain, TrkC-T1, is a determinant of the corticofugal projection neuron (CFuPN) fate. Here, we show that the finely tuned balance between TrkC-T1 and the better known, kinase domain-containing isoform, TrkC-TK+, is cell type-specific in the developing cortex and established through the antagonistic actions of two RNA-binding proteins, Srsf1 and Elavl1. Moreover, our data show that Srsf1 promotes the CFuPN fate and Elavl1 promotes the callosal projection neuron (CPN) fate in vivo via regulating the distinct ratios of TrkC-T1 to TrkC-TK+. Taken together, we connect spatio-temporal expression of Srsf1 and Elavl1 in the developing neocortex with the regulation of TrkC alternative splicing and transcript stability and neuronal fate choice, thus adding to the mechanistic and functional understanding of alternative splicing in vivo.
Collapse
Affiliation(s)
- A Ioana Weber
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 6, 14195, Berlin, Germany
| | - Srinivas Parthasarathy
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Ekaterina Borisova
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| | - Ekaterina Epifanova
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Marco Preußner
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 6, 14195, Berlin, Germany
| | - Alexandra Rusanova
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634009, Tomsk, Russia
| | - Mateusz C Ambrozkiewicz
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Paraskevi Bessa
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Andrew G Newman
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Lisa Müller
- Heinrich Heine Universität Düsseldorf, Institute of Virology, Medical Faculty, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Heiner Schaal
- Heinrich Heine Universität Düsseldorf, Institute of Virology, Medical Faculty, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Florian Heyd
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Takustr. 6, 14195, Berlin, Germany
| | - Victor Tarabykin
- Charité Universitätsmedizin Berlin, Institute of Cell Biology and Neurobiology, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, 603950, Nizhny Novgorod Oblast, Russia
| |
Collapse
|
13
|
Saha S, Jungas TT, Ohayon D, Audouard C, Ye T, Fawal MA, Davy A. Dihydrofolate reductase activity controls neurogenic transitions in the developing neocortex. Development 2023; 150:dev201696. [PMID: 37665322 DOI: 10.1242/dev.201696] [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: 02/11/2023] [Accepted: 08/01/2023] [Indexed: 09/05/2023]
Abstract
One-carbon/folate (1C) metabolism supplies methyl groups required for DNA and histone methylation, and is involved in the maintenance of self-renewal in stem cells. Dihydrofolate reductase (DHFR), a key enzyme in 1C metabolism, is highly expressed in human and mouse neural progenitors at the early stages of neocortical development. Here, we have investigated the role of DHFR in the developing neocortex and report that reducing its activity in human neural organoids and mouse embryonic neocortex accelerates indirect neurogenesis, thereby affecting neuronal composition of the neocortex. Furthermore, we show that decreasing DHFR activity in neural progenitors leads to a reduction in one-carbon/folate metabolites and correlates with modifications of H3K4me3 levels. Our findings reveal an unanticipated role for DHFR in controlling specific steps of neocortex development and indicate that variations in 1C metabolic cues impact cell fate transitions.
Collapse
Affiliation(s)
- Sulov Saha
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Thomas T Jungas
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - David Ohayon
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Christophe Audouard
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Tao Ye
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS UMR7104, Université de Strasbourg, 1 rue Laurent Fries, 67404 Illkirch, France
| | - Mohamad-Ali Fawal
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| | - Alice Davy
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062 Toulouse, France
| |
Collapse
|
14
|
Polenghi M, Taverna E. Intracellular traffic and polarity in brain development. Front Neurosci 2023; 17:1172016. [PMID: 37859764 PMCID: PMC10583573 DOI: 10.3389/fnins.2023.1172016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/31/2023] [Indexed: 10/21/2023] Open
Abstract
Neurons forming the human brain are generated during embryonic development by neural stem and progenitor cells via a process called neurogenesis. A crucial feature contributing to neural stem cell morphological and functional heterogeneity is cell polarity, defined as asymmetric distribution of cellular components. Cell polarity is built and maintained thanks to the interplay between polarity proteins and polarity-generating organelles, such as the endoplasmic reticulum (ER) and the Golgi apparatus (GA). ER and GA affect the distribution of membrane components and work as a hub where glycans are added to nascent proteins and lipids. In the last decades our knowledge on the role of polarity in neural stem and progenitor cells have increased tremendously. However, the role of traffic and associated glycosylation in neural stem and progenitor cells is still relatively underexplored. In this review, we discuss the link between cell polarity, architecture, identity and intracellular traffic, and highlight how studies on neurons have shaped our knowledge and conceptual framework on traffic and polarity. We will then conclude by discussing how a group of rare diseases, called congenital disorders of glycosylation (CDG) offers the unique opportunity to study the contribution of traffic and glycosylation in the context of neurodevelopment.
Collapse
|
15
|
Martins M, Oliveira AR, Martins S, Vieira JP, Perdigão P, Fernandes AR, de Almeida LP, Palma PJ, Sequeira DB, Santos JMM, Duque F, Oliveira G, Cardoso AL, Peça J, Seabra CM. A Novel Genetic Variant in MBD5 Associated with Severe Epilepsy and Intellectual Disability: Potential Implications on Neural Primary Cilia. Int J Mol Sci 2023; 24:12603. [PMID: 37628781 PMCID: PMC10454663 DOI: 10.3390/ijms241612603] [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: 07/14/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Disruptions in the MBD5 gene have been linked with an array of clinical features such as global developmental delay, intellectual disability, autistic-like symptoms, and seizures, through unclear mechanisms. MBD5 haploinsufficiency has been associated with the disruption of primary cilium-related processes during early cortical development, and this has been reported in many neurodevelopmental disorders. In this study, we describe the clinical history of a 12-year-old child harboring a novel MBD5 rare variant and presenting psychomotor delay and seizures. To investigate the impact of MBD5 haploinsufficiency on neural primary cilia, we established a novel patient-derived cell line and used CRISPR-Cas9 technology to create an isogenic control. The patient-derived neural progenitor cells revealed a decrease in the length of primary cilia and in the total number of ciliated cells. This study paves the way to understanding the impact of MBD5 haploinsufficiency in brain development through its potential impact on neural primary cilia.
Collapse
Affiliation(s)
- Mariana Martins
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Rafaela Oliveira
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Solange Martins
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - José Pedro Vieira
- Neuropediatrics Unit, Central Lisbon Hospital Center, 1169-045 Lisbon, Portugal
| | - Pedro Perdigão
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Ana Rita Fernandes
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Paulo Jorge Palma
- Institute of Endodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Center for Innovation and Research in Oral Sciences (CIROS), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Diana Bela Sequeira
- Institute of Endodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Center for Innovation and Research in Oral Sciences (CIROS), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - João Miguel Marques Santos
- Institute of Endodontics, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Center for Innovation and Research in Oral Sciences (CIROS), Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
| | - Frederico Duque
- University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, 3000-602 Coimbra, Portugal
- Child Developmental Center and Research and Clinical Training Center, Pediatric Hospital, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-602 Coimbra, Portugal
| | - Guiomar Oliveira
- University Clinic of Pediatrics, Faculty of Medicine, University of Coimbra, 3000-602 Coimbra, Portugal
- Child Developmental Center and Research and Clinical Training Center, Pediatric Hospital, Centro Hospitalar e Universitário de Coimbra (CHUC), 3000-602 Coimbra, Portugal
| | - Ana Luísa Cardoso
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| | - João Peça
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3000-456 Coimbra, Portugal
| | - Catarina Morais Seabra
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
16
|
Alcaide Martin A, Mayerl S. Local Thyroid Hormone Action in Brain Development. Int J Mol Sci 2023; 24:12352. [PMID: 37569727 PMCID: PMC10418487 DOI: 10.3390/ijms241512352] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/13/2023] Open
Abstract
Proper brain development essentially depends on the timed availability of sufficient amounts of thyroid hormone (TH). This, in turn, necessitates a tightly regulated expression of TH signaling components such as TH transporters, deiodinases, and TH receptors in a brain region- and cell-specific manner from early developmental stages onwards. Abnormal TH levels during critical stages, as well as mutations in TH signaling components that alter the global and/or local thyroidal state, result in detrimental consequences for brain development and neurological functions that involve alterations in central neurotransmitter systems. Thus, the question as to how TH signaling is implicated in the development and maturation of different neurotransmitter and neuromodulator systems has gained increasing attention. In this review, we first summarize the current knowledge on the regulation of TH signaling components during brain development. We then present recent advances in our understanding on how altered TH signaling compromises the development of cortical glutamatergic neurons, inhibitory GABAergic interneurons, cholinergic and dopaminergic neurons. Thereby, we highlight novel mechanistic insights and point out open questions in this evolving research field.
Collapse
Affiliation(s)
| | - Steffen Mayerl
- Department of Endocrinology Diabetes & Metabolism, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany
| |
Collapse
|
17
|
Gkini V, Namba T. Glutaminolysis and the Control of Neural Progenitors in Neocortical Development and Evolution. Neuroscientist 2023; 29:177-189. [PMID: 35057642 PMCID: PMC10018057 DOI: 10.1177/10738584211069060] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple types of neural progenitor cells (NPCs) contribute to the development of the neocortex, a brain region responsible for our higher cognitive abilities. Proliferative capacity of NPCs varies among NPC types, developmental stages, and species. The higher proliferative capacity of NPCs in the developing human neocortex is thought to be a major contributing factor why humans have the most expanded neocortex within primates. Recent studies have shed light on the importance of cell metabolism in the neocortical NPC proliferative capacity. Specifically, glutaminolysis, a metabolic pathway that converts glutamine to glutamate and then to α-ketoglutarate, has been shown to play a critical role in human NPCs, both in apical and basal progenitors. In this review, we summarize our current knowledge of NPC metabolism, focusing especially on glutaminolysis, and discuss the role of NPC metabolism in neocortical development, evolution, and neurodevelopmental disorders, providing a broader perspective on a newly emerging research field.
Collapse
Affiliation(s)
- Vasiliki Gkini
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
- Takashi Namba, Neuroscience Center, HiLIFE
— Helsinki Institute of Life Science, University of Helsinki, PO 63,
Haartmaninkatu 8, Helsinki 00014, Finland.
| |
Collapse
|
18
|
Zhang D, Eguchi N, Okazaki S, Sora I, Hishimoto A. Telencephalon Organoids Derived from an Individual with ADHD Show Altered Neurodevelopment of Early Cortical Layer Structure. Stem Cell Rev Rep 2023:10.1007/s12015-023-10519-z. [PMID: 36872412 PMCID: PMC10366301 DOI: 10.1007/s12015-023-10519-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/07/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder that occurs in early childhood and can persist to adulthood. It can affect many aspects of a patient's daily life, so it is necessary to explore the mechanism and pathological alterations. For this purpose, we applied induced pluripotent stem cell (iPSC)-derived telencephalon organoids to recapitulate the alterations occurring in the early cerebral cortex of ADHD patients. We found that telencephalon organoids of ADHD showed less growth of layer structures than control-derived organoids. On day 35 of differentiation, the thinner cortex layer structures of ADHD-derived organoids contained more neurons than those of control-derived organoids. Furthermore, ADHD-derived organoids showed a decrease in cell proliferation during development from day 35 to 56. On day 56 of differentiation, there was a significant difference in the proportion of symmetric and asymmetric cell division between the ADHD and control groups. In addition, we observed increased cell apoptosis in ADHD during early development. These results show alterations in the characteristics of neural stem cells and the formation of layer structures, which might indicate key roles in the pathogenesis of ADHD. Our organoids exhibit the cortical developmental alterations observed in neuroimaging studies, providing an experimental foundation for understanding the pathological mechanisms of ADHD.
Collapse
Affiliation(s)
- Danmeng Zhang
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriomi Eguchi
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satoshi Okazaki
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ichiro Sora
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Akitoyo Hishimoto
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
19
|
Vaid S, Heikinheimo O, Namba T. Embryonic mouse medial neocortex as a model system for studying the radial glial scaffold in fetal human neocortex. J Neural Transm (Vienna) 2023; 130:185-194. [PMID: 36450874 PMCID: PMC10033555 DOI: 10.1007/s00702-022-02570-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
Neocortex is the evolutionarily newest region in the brain, and is a structure with diversified size and morphology among mammalian species. Humans have the biggest neocortex compared to the body size, and their neocortex has many foldings, that is, gyri and sulci. Despite the recent methodological advances in in vitro models such as cerebral organoids, mice have been continuously used as a model system for studying human neocortical development because of the accessibility and practicality of in vivo gene manipulation. The commonly studied neocortical region, the lateral neocortex, generally recapitulates the developmental process of the human neocortex, however, there are several important factors missing in the lateral neocortex. First, basal (outer) radial glia (bRG), which are the main cell type providing the radial scaffold to the migrating neurons in the fetal human neocortex, are very few in the mouse lateral neocortex, thus the radial glial scaffold is different from the fetal human neocortex. Second, as a consequence of the difference in the radial glial scaffold, migrating neurons might exhibit different migratory behavior and thus distribution. To overcome those problems, we propose the mouse medial neocortex, where we have earlier revealed an abundance of bRG similar to the fetal human neocortex, as an alternative model system. We found that similar to the fetal human neocortex, the radial glial scaffold, neuronal migration and neuronal distribution are tangentially scattered in the mouse medial neocortex. Taken together, the embryonic mouse medial neocortex could be a suitable and accessible in vivo model system to study human neocortical development and its pathogenesis.
Collapse
Affiliation(s)
- Samir Vaid
- Department of Basic Neurosciences, University of Geneva, 1211, Geneva, Switzerland
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, P.O. 140, 00029, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, P.O. 63, 00014, Helsinki, Finland.
| |
Collapse
|
20
|
Li S, Hannenhalli S, Ovcharenko I. De novo human brain enhancers created by single-nucleotide mutations. SCIENCE ADVANCES 2023; 9:eadd2911. [PMID: 36791193 PMCID: PMC9931207 DOI: 10.1126/sciadv.add2911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 01/12/2023] [Indexed: 05/30/2023]
Abstract
Advanced human cognition is attributed to increased neocortex size and complexity, but the underlying evolutionary and regulatory mechanisms are largely unknown. Using human and macaque embryonic neocortical H3K27ac data coupled with a deep learning model of enhancers, we identified ~4000 enhancer gains in humans, which, per our model, can often be attributed to single-nucleotide essential mutations. Our analyses suggest that functional gains in embryonic brain development are associated with de novo enhancers whose putative target genes exhibit increased expression in progenitor cells and interneurons and partake in critical neural developmental processes. Essential mutations alter enhancer activity through altered binding of key transcription factors (TFs) of embryonic neocortex, including ISL1, POU3F2, PITX1/2, and several SOX TFs, and are associated with central nervous system disorders. Overall, our results suggest that essential mutations lead to gain of embryonic neocortex enhancers, which orchestrate expression of genes involved in critical developmental processes associated with human cognition.
Collapse
Affiliation(s)
- Shan Li
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ivan Ovcharenko
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Ritchie FD, Lizarraga SB. The role of histone methyltransferases in neurocognitive disorders associated with brain size abnormalities. Front Neurosci 2023; 17:989109. [PMID: 36845425 PMCID: PMC9950662 DOI: 10.3389/fnins.2023.989109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 01/17/2023] [Indexed: 02/12/2023] Open
Abstract
Brain size is controlled by several factors during neuronal development, including neural progenitor proliferation, neuronal arborization, gliogenesis, cell death, and synaptogenesis. Multiple neurodevelopmental disorders have co-morbid brain size abnormalities, such as microcephaly and macrocephaly. Mutations in histone methyltransferases that modify histone H3 on Lysine 36 and Lysine 4 (H3K36 and H3K4) have been identified in neurodevelopmental disorders involving both microcephaly and macrocephaly. H3K36 and H3K4 methylation are both associated with transcriptional activation and are proposed to sterically hinder the repressive activity of the Polycomb Repressor Complex 2 (PRC2). During neuronal development, tri-methylation of H3K27 (H3K27me3) by PRC2 leads to genome wide transcriptional repression of genes that regulate cell fate transitions and neuronal arborization. Here we provide a review of neurodevelopmental processes and disorders associated with H3K36 and H3K4 histone methyltransferases, with emphasis on processes that contribute to brain size abnormalities. Additionally, we discuss how the counteracting activities of H3K36 and H3K4 modifying enzymes vs. PRC2 could contribute to brain size abnormalities which is an underexplored mechanism in relation to brain size control.
Collapse
|
22
|
Lafferty MJ, Aygün N, Patel NK, Krupa O, Liang D, Wolter JM, Geschwind DH, de la Torre-Ubieta L, Stein JL. MicroRNA-eQTLs in the developing human neocortex link miR-4707-3p expression to brain size. eLife 2023; 12:e79488. [PMID: 36629315 PMCID: PMC9859047 DOI: 10.7554/elife.79488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 01/10/2023] [Indexed: 01/12/2023] Open
Abstract
Expression quantitative trait loci (eQTL) data have proven important for linking non-coding loci to protein-coding genes. But eQTL studies rarely measure microRNAs (miRNAs), small non-coding RNAs known to play a role in human brain development and neurogenesis. Here, we performed small-RNA sequencing across 212 mid-gestation human neocortical tissue samples, measured 907 expressed miRNAs, discovering 111 of which were novel, and identified 85 local-miRNA-eQTLs. Colocalization of miRNA-eQTLs with GWAS summary statistics yielded one robust colocalization of miR-4707-3p expression with educational attainment and brain size phenotypes, where the miRNA expression increasing allele was associated with decreased brain size. Exogenous expression of miR-4707-3p in primary human neural progenitor cells decreased expression of predicted targets and increased cell proliferation, indicating miR-4707-3p modulates progenitor gene regulation and cell fate decisions. Integrating miRNA-eQTLs with existing GWAS yielded evidence of a miRNA that may influence human brain size and function via modulation of neocortical brain development.
Collapse
Affiliation(s)
- Michael J Lafferty
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Nil Aygün
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Niyanta K Patel
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Oleh Krupa
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Dan Liang
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
| | - Justin M Wolter
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel HillChapel HillUnited States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel HillChapel HillUnited States
| | - Daniel H Geschwind
- Neurogenetics Program, Department of Neurology, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Luis de la Torre-Ubieta
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel HillChapel HillUnited States
- UNC Neuroscience Center, University of North Carolina at Chapel HillChapel HillUnited States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel HillChapel HillUnited States
| |
Collapse
|
23
|
Levchenko A, Gusev F, Rogaev E. The evolutionary origin of psychosis. Front Psychiatry 2023; 14:1115929. [PMID: 36741116 PMCID: PMC9894884 DOI: 10.3389/fpsyt.2023.1115929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
Imagination, the driving force of creativity, and primary psychosis are human-specific, since we do not observe behaviors in other species that would convincingly suggest they possess the same traits. Both these traits have been linked to the function of the prefrontal cortex, which is the most evolutionarily novel region of the human brain. A number of evolutionarily novel genetic and epigenetic changes that determine the human brain-specific structure and function have been discovered in recent years. Among them are genomic loci subjected to increased rates of single nucleotide substitutions in humans, called human accelerated regions. These mostly regulatory regions are involved in brain development and sometimes contain genetic variants that confer a risk for schizophrenia. On the other hand, neuroimaging data suggest that mind wandering and related phenomena (as a proxy of imagination) are in many ways similar to rapid eye movement dreaming, a function also present in non-human species. Furthermore, both functions are similar to psychosis in several ways: for example, the same brain areas are activated both in dreams and visual hallucinations. In the present Perspective we hypothesize that imagination is an evolutionary adaptation of dreaming, while primary psychosis results from deficient control by higher-order brain areas over imagination. In the light of this, human accelerated regions might be one of the key drivers in evolution of human imagination and the pathogenesis of psychotic disorders.
Collapse
Affiliation(s)
- Anastasia Levchenko
- Institute of Translational Biomedicine, Saint Petersburg State University, Saint Petersburg, Russia
| | - Fedor Gusev
- Center for Genetics and Life Sciences, Department of Genetics, Sirius University of Science and Technology, Sochi, Russia.,Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Evgeny Rogaev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia.,Department of Psychiatry, UMass Chan Medical School, Shrewsbury, MA, United States
| |
Collapse
|
24
|
Degl’Innocenti E, Dell’Anno MT. Human and mouse cortical astrocytes: a comparative view from development to morphological and functional characterization. Front Neuroanat 2023; 17:1130729. [PMID: 37139179 PMCID: PMC10150887 DOI: 10.3389/fnana.2023.1130729] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
The vision of astroglia as a bare scaffold to neuronal circuitry has been largely overturned. Astrocytes exert a neurotrophic function, but also take active part in supporting synaptic transmission and in calibrating blood circulation. Many aspects of their functioning have been unveiled from studies conducted in murine models, however evidence is showing many differences between mouse and human astrocytes starting from their development and encompassing morphological, transcriptomic and physiological variations when they achieve complete maturation. The evolutionary race toward superior cognitive abilities unique to humans has drastically impacted neocortex structure and, together with neuronal circuitry, astrocytes have also been affected with the acquisition of species-specific properties. In this review, we summarize diversities between murine and human astroglia, with a specific focus on neocortex, in a panoramic view that starts with their developmental origin to include all structural and molecular differences that mark the uniqueness of human astrocytes.
Collapse
Affiliation(s)
- Elisa Degl’Innocenti
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Maria Teresa Dell’Anno
- Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy
- *Correspondence: Maria Teresa Dell’Anno,
| |
Collapse
|
25
|
Lemaitre H, Le Guen Y, Tilot AK, Stein JL, Philippe C, Mangin JF, Fisher SE, Frouin V. Genetic variations within human gained enhancer elements affect human brain sulcal morphology. Neuroimage 2023; 265:119773. [PMID: 36442731 DOI: 10.1016/j.neuroimage.2022.119773] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 10/07/2022] [Accepted: 11/24/2022] [Indexed: 11/26/2022] Open
Abstract
The expansion of the cerebral cortex is one of the most distinctive changes in the evolution of the human brain. Cortical expansion and related increases in cortical folding may have contributed to emergence of our capacities for high-order cognitive abilities. Molecular analysis of humans, archaic hominins, and non-human primates has allowed identification of chromosomal regions showing evolutionary changes at different points of our phylogenetic history. In this study, we assessed the contributions of genomic annotations spanning 30 million years to human sulcal morphology measured via MRI in more than 18,000 participants from the UK Biobank. We found that variation within brain-expressed human gained enhancers, regulatory genetic elements that emerged since our last common ancestor with Old World monkeys, explained more trait heritability than expected for the left and right calloso-marginal posterior fissures and the right central sulcus. Intriguingly, these are sulci that have been previously linked to the evolution of locomotion in primates and later on bipedalism in our hominin ancestors.
Collapse
Affiliation(s)
- Herve Lemaitre
- Institut des Maladies Neurodégénératives, CNRS UMR 5293, Université de bordeaux, Centre Broca Nouvelle-Aquitaine, Bordeaux, France.
| | - Yann Le Guen
- Université Paris-Saclay, CEA, CNRS, Neurospin, Baobab UMR 9027, Gif-sur-Yvette, France
| | - Amanda K Tilot
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands
| | - Jason L Stein
- Department of Genetics and the UNC Neuroscience Center, UNC-Chapel Hill, Chapel Hill, NC, United States of America
| | - Cathy Philippe
- Université Paris-Saclay, CEA, CNRS, Neurospin, Baobab UMR 9027, Gif-sur-Yvette, France
| | - Jean-François Mangin
- Université Paris-Saclay, CEA, CNRS, Neurospin, Baobab UMR 9027, Gif-sur-Yvette, France
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, Netherlands
| | - Vincent Frouin
- Université Paris-Saclay, CEA, CNRS, Neurospin, Baobab UMR 9027, Gif-sur-Yvette, France
| |
Collapse
|
26
|
Alkailani MI, Aittaleb M, Tissir F. WNT signaling at the intersection between neurogenesis and brain tumorigenesis. Front Mol Neurosci 2022; 15:1017568. [PMID: 36267699 PMCID: PMC9577257 DOI: 10.3389/fnmol.2022.1017568] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
Neurogenesis and tumorigenesis share signaling molecules/pathways involved in cell proliferation, differentiation, migration, and death. Self-renewal of neural stem cells is a tightly regulated process that secures the accuracy of cell division and eliminates cells that undergo mitotic errors. Abnormalities in the molecular mechanisms controlling this process can trigger aneuploidy and genome instability, leading to neoplastic transformation. Mutations that affect cell adhesion, polarity, or migration enhance the invasive potential and favor the progression of tumors. Here, we review recent evidence of the WNT pathway’s involvement in both neurogenesis and tumorigenesis and discuss the experimental progress on therapeutic opportunities targeting components of this pathway.
Collapse
Affiliation(s)
- Maisa I. Alkailani
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Mohamed Aittaleb
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Fadel Tissir
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
- Institute of Neuroscience, Université catholique de Louvain, Brussels, Belgium
- *Correspondence: Fadel Tissir,
| |
Collapse
|
27
|
Park SHE, Ortiz AK, Konopka G. Corticogenesis across species at single-cell resolution. Dev Neurobiol 2022; 82:517-532. [PMID: 35932776 PMCID: PMC9481703 DOI: 10.1002/dneu.22896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 07/11/2022] [Accepted: 07/22/2022] [Indexed: 11/07/2022]
Abstract
The neocortex (or pallium) consists of diverse cell types that are organized in a highly species-specific manner under strict spatiotemporal control during development. Many of the cell types are present transiently throughout development but contribute to permanent species-specific cortical features that are acquired through evolution. Therefore, capturing cell type-specific biological information has always been an important quest in the field of neurodevelopment. The progress in achieving fine cellular resolution has been slow due to technical challenges. However, with recent advancements in single-cell and multi-omics technologies, many laboratories have begun to successfully interrogate cellular and molecular mechanisms driving corticogenesis at single-cell resolution. In this review, we provide summarized results from many primary publications and several in-depth review articles that utilize or address single-cell genomics techniques to understand important topics, such as cellular and molecular mechanisms governing cortical progenitor proliferation, cell lineage progression, neuronal specification, and arealization, across multiple gyrencephalic (i.e., human and non-human primates) and lissencephalic species (i.e., mouse, reptiles, and songbirds). We also examine findings from recent studies involving epigenomic and posttranscriptional regulation of corticogenesis. In the discussion section, we provide our insights on the challenges the field currently faces as well as promising future applications of single cell technologies.
Collapse
Affiliation(s)
- Seon Hye E Park
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ana K Ortiz
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
28
|
Drugs and Endogenous Factors as Protagonists in Neurogenic Stimulation. Stem Cell Rev Rep 2022; 18:2852-2871. [PMID: 35962176 DOI: 10.1007/s12015-022-10423-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 10/15/2022]
Abstract
Neurogenesis is a biological process characterized by new neurons formation from stem cells. For decades, it was believed that neurons only multiplied during development and in the postnatal period but the discovery of neural stem cells (NSCs) in mature brain promoted a revolution in neuroscience field. In mammals, neurogenesis consists of migration, differentiation, maturation, as well as functional integration of newborn cells into the pre-existing neuronal circuit. Actually, NSC density drops significantly after the first stages of development, however in specific places in the brain, called neurogenic niches, some of these cells retain their ability to generate new neurons and glial cells in adulthood. The subgranular (SGZ), and the subventricular zones (SVZ) are examples of regions where the neurogenesis process occurs in the mature brain. There, the potential of NSCs to produce new neurons has been explored by new advanced methodologies and in neuroscience for the treatment of brain damage and/or degeneration. Based on that, this review highlights endogenous factors and drugs capable of stimulating neurogenesis, as well as the perspectives for the use of NSCs for neurological and neurodegenerative diseases.
Collapse
|
29
|
Zhu X, Guo Y, Chu C, Liu D, Duan K, Yin Y, Si C, Kang Y, Yao J, Du X, Li J, Zhao S, Ai Z, Zhu Q, Ji W, Niu Y, Li T. BRN2 as a key gene drives the early primate telencephalon development. SCIENCE ADVANCES 2022; 8:eabl7263. [PMID: 35245119 PMCID: PMC8896791 DOI: 10.1126/sciadv.abl7263] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
Evolutionary mutations in primate-specific genes drove primate cortex expansion. However, whether conserved genes with previously unidentified functions also play a key role in primate brain expansion remains unknown. Here, we focus on BRN2 (POU3F2), a gene encoding a neural transcription factor commonly expressed in both primates and mice. Compared to the limited effects on mouse brain development, BRN2 biallelic knockout in cynomolgus monkeys (Macaca fascicularis) is lethal before midgestation. Histology analysis and single-cell transcriptome show that BRN2 deficiency decreases RGC expansion, induces precocious differentiation, and alters the trajectory of neurogenesis in the telencephalon. BRN2, serving as an upstream factor, controls specification and differentiation of ganglionic eminences. In addition, we identified the conserved function of BRN2 in cynomolgus monkeys to human RGCs. BRN2 may function by directly regulating SOX2 and STAT3 and maintaining HOPX. Our findings reveal a previously unknown mechanism that BRN2, a conserved gene, drives early primate telencephalon development by gaining novel mechanistic functions.
Collapse
Affiliation(s)
- Xiaoqing Zhu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yicheng Guo
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Chu Chu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Dahai Liu
- Department of Basic Medicine and Biomedical Engineering, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong 528000, China
| | - Kui Duan
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Chenyang Si
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yu Kang
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Junjun Yao
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Xuewei Du
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Junliang Li
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Shumei Zhao
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Zongyong Ai
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Qingyuan Zhu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Yuyu Niu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China
| |
Collapse
|
30
|
Girskis KM, Stergachis AB, DeGennaro EM, Doan RN, Qian X, Johnson MB, Wang PP, Sejourne GM, Nagy MA, Pollina EA, Sousa AMM, Shin T, Kenny CJ, Scotellaro JL, Debo BM, Gonzalez DM, Rento LM, Yeh RC, Song JHT, Beaudin M, Fan J, Kharchenko PV, Sestan N, Greenberg ME, Walsh CA. Rewiring of human neurodevelopmental gene regulatory programs by human accelerated regions. Neuron 2021; 109:3239-3251.e7. [PMID: 34478631 DOI: 10.1016/j.neuron.2021.08.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/02/2021] [Accepted: 08/06/2021] [Indexed: 01/21/2023]
Abstract
Human accelerated regions (HARs) are the fastest-evolving regions of the human genome, and many are hypothesized to function as regulatory elements that drive human-specific gene regulatory programs. We interrogate the in vitro enhancer activity and in vivo epigenetic landscape of more than 3,100 HARs during human neurodevelopment, demonstrating that many HARs appear to act as neurodevelopmental enhancers and that sequence divergence at HARs has largely augmented their neuronal enhancer activity. Furthermore, we demonstrate PPP1R17 to be a putative HAR-regulated gene that has undergone remarkable rewiring of its cell type and developmental expression patterns between non-primates and primates and between non-human primates and humans. Finally, we show that PPP1R17 slows neural progenitor cell cycle progression, paralleling the cell cycle length increase seen predominantly in primate and especially human neurodevelopment. Our findings establish HARs as key components in rewiring human-specific neurodevelopmental gene regulatory programs and provide an integrated resource to study enhancer activity of specific HARs.
Collapse
Affiliation(s)
- Kelly M Girskis
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Andrew B Stergachis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Ellen M DeGennaro
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan N Doan
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xuyu Qian
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew B Johnson
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Peter P Wang
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gabrielle M Sejourne
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - M Aurel Nagy
- Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Pollina
- Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - André M M Sousa
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Taehwan Shin
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Program in Biological and Biomedical Sciences, Harvard Medical School, Boston MA, USA
| | - Connor J Kenny
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Julia L Scotellaro
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brian M Debo
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Dilenny M Gonzalez
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lariza M Rento
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rebecca C Yeh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Janet H T Song
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc Beaudin
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jean Fan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter V Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Nenad Sestan
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Michael E Greenberg
- Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Kalebic N, Namba T. Inheritance and flexibility of cell polarity: a clue for understanding human brain development and evolution. Development 2021; 148:272121. [PMID: 34499710 PMCID: PMC8451944 DOI: 10.1242/dev.199417] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cell polarity is fundamentally important for understanding brain development. Here, we hypothesize that the inheritance and flexibility of cell polarity during neocortex development could be implicated in neocortical evolutionary expansion. Molecular and morphological features of cell polarity may be inherited from one type of progenitor cell to the other and finally transmitted to neurons. Furthermore, key cell types, such as basal progenitors and neurons, exhibit a highly flexible polarity. We suggest that both inheritance and flexibility of cell polarity are implicated in the amplification of basal progenitors and tangential dispersion of neurons, which are key features of the evolutionary expansion of the neocortex. Summary: We suggest that the inheritance and flexibility of cell polarity are implicated in the evolutionary expansion of the developing neocortex by promoting the amplification of neural progenitors and tangential migration of neurons.
Collapse
Affiliation(s)
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
32
|
How neural stem cells contribute to neocortex development. Biochem Soc Trans 2021; 49:1997-2006. [PMID: 34397081 PMCID: PMC8589419 DOI: 10.1042/bst20200923] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/15/2021] [Accepted: 07/29/2021] [Indexed: 11/30/2022]
Abstract
The mammalian neocortex is the seat of higher cognitive functions, such as thinking and language in human. A hallmark of the neocortex are the cortical neurons, which are generated from divisions of neural progenitor cells (NPCs) during development, and which constitute a key feature of the well-organized layered structure of the neocortex. Proper formation of neocortex structure requires an orchestrated cellular behavior of different cortical NPCs during development, especially during the process of cortical neurogenesis. Here, we review the great diversity of NPCs and their contribution to the development of the neocortex. First, we review the categorization of NPCs into different classes and types based on their cell biological features, and discuss recent advances in characterizing marker expression and cell polarity features in the different types of NPCs. Second, we review the different modes of cell divisions that NPCs undergo and discuss the importance of the balance between proliferation and differentiation of NPCs in neocortical development. Third, we review the different proliferative capacities among different NPC types and among the same type of NPC in different mammalian species. Dissecting the differences between NPC types and differences among mammalian species is beneficial to further understand the development and the evolutionary expansion of the neocortex and may open up new therapeutic avenues for neurodevelopmental and psychiatric disorders.
Collapse
|
33
|
Micali N, Kim SK, Diaz-Bustamante M, Stein-O'Brien G, Seo S, Shin JH, Rash BG, Ma S, Wang Y, Olivares NA, Arellano JI, Maynard KR, Fertig EJ, Cross AJ, Bürli RW, Brandon NJ, Weinberger DR, Chenoweth JG, Hoeppner DJ, Sestan N, Rakic P, Colantuoni C, McKay RD. Variation of Human Neural Stem Cells Generating Organizer States In Vitro before Committing to Cortical Excitatory or Inhibitory Neuronal Fates. Cell Rep 2021; 31:107599. [PMID: 32375049 PMCID: PMC7357345 DOI: 10.1016/j.celrep.2020.107599] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 10/22/2019] [Accepted: 04/10/2020] [Indexed: 11/06/2022] Open
Abstract
Better understanding of the progression of neural stem cells (NSCs) in the developing cerebral cortex is important for modeling neurogenesis and defining the pathogenesis of neuropsychiatric disorders. Here, we use RNA sequencing, cell imaging, and lineage tracing of mouse and human in vitro NSCs and monkey brain sections to model the generation of cortical neuronal fates. We show that conserved signaling mechanisms regulate the acute transition from proliferative NSCs to committed glutamatergic excitatory neurons. As human telencephalic NSCs develop from pluripotency in vitro, they transition through organizer states that spatially pattern the cortex before generating glutamatergic precursor fates. NSCs derived from multiple human pluripotent lines vary in these early patterning states, leading differentially to dorsal or ventral telencephalic fates. This work furthers systematic analyses of the earliest patterning events that generate the major neuronal trajectories of the human telencephalon. Micali et al. report that human telencephalic NSCs in vitro transition through the organizer states that pattern the neocortex. Human pluripotent lines vary in organizer formation, generating divergent neuronal differentiation trajectories biased toward dorsal or ventral telencephalic fates and opening further analysis of the earliest cortical specification events.
Collapse
Affiliation(s)
- Nicola Micali
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Suel-Kee Kim
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Genevieve Stein-O'Brien
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Seungmae Seo
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA
| | - Joo-Heon Shin
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA
| | - Brian G Rash
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shaojie Ma
- Departments of Comparative Medicine, Genetics, and Psychiatry, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yanhong Wang
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA
| | - Nicolas A Olivares
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA
| | - Jon I Arellano
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA
| | - Elana J Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Applied Mathematics and Statistics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Alan J Cross
- AstraZeneca Neuroscience, IMED Biotech Unit, R&D, Boston, MA 024515, USA
| | - Roland W Bürli
- AstraZeneca Neuroscience, IMED Biotech Unit, R&D, Boston, MA 024515, USA
| | - Nicholas J Brandon
- AstraZeneca Neuroscience, IMED Biotech Unit, R&D, Boston, MA 024515, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Joshua G Chenoweth
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA
| | - Daniel J Hoeppner
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA; Astellas Research Institute of America, 3565 General Atomics Ct., Ste. 200, San Diego, CA 92121, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA; Departments of Comparative Medicine, Genetics, and Psychiatry, Yale School of Medicine, New Haven, CT 06520, USA; Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA; Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Carlo Colantuoni
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Ronald D McKay
- Lieber Institute for Brain Development, 855 North Wolfe St., Baltimore, MD 21205, USA.
| |
Collapse
|
34
|
An Evolved Human-specific Epigenetic Mechanism for Cortical Expansion and Gyrification. Neurosci Bull 2021; 37:1370-1372. [PMID: 34076853 DOI: 10.1007/s12264-021-00719-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/05/2021] [Indexed: 10/21/2022] Open
|
35
|
Nucleocytoplasmic transport of the RNA-binding protein CELF2 regulates neural stem cell fates. Cell Rep 2021; 35:109226. [PMID: 34107259 DOI: 10.1016/j.celrep.2021.109226] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/23/2021] [Accepted: 05/13/2021] [Indexed: 01/12/2023] Open
Abstract
The development of the cerebral cortex requires balanced expansion and differentiation of neural stem/progenitor cells (NPCs), which rely on precise regulation of gene expression. Because NPCs often exhibit transcriptional priming of cell-fate-determination genes, the ultimate output of these genes for fate decisions must be carefully controlled in a timely fashion at the post-transcriptional level, but how that is achieved is poorly understood. Here, we report that de novo missense variants in an RNA-binding protein CELF2 cause human cortical malformations and perturb NPC fate decisions in mice by disrupting CELF2 nucleocytoplasmic transport. In self-renewing NPCs, CELF2 resides in the cytoplasm, where it represses mRNAs encoding cell fate regulators and neurodevelopmental disorder-related factors. The translocation of CELF2 into the nucleus releases mRNA for translation and thereby triggers NPC differentiation. Our results reveal that CELF2 translocation between subcellular compartments orchestrates mRNA at the translational level to instruct cell fates in cortical development.
Collapse
|
36
|
When Good Kinases Go Rogue: GSK3, p38 MAPK and CDKs as Therapeutic Targets for Alzheimer's and Huntington's Disease. Int J Mol Sci 2021; 22:ijms22115911. [PMID: 34072862 PMCID: PMC8199025 DOI: 10.3390/ijms22115911] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is a mostly sporadic brain disorder characterized by cognitive decline resulting from selective neurodegeneration in the hippocampus and cerebral cortex whereas Huntington's disease (HD) is a monogenic inherited disorder characterized by motor abnormalities and psychiatric disturbances resulting from selective neurodegeneration in the striatum. Although there have been numerous clinical trials for these diseases, they have been unsuccessful. Research conducted over the past three decades by a large number of laboratories has demonstrated that abnormal actions of common kinases play a key role in the pathogenesis of both AD and HD as well as several other neurodegenerative diseases. Prominent among these kinases are glycogen synthase kinase (GSK3), p38 mitogen-activated protein kinase (MAPK) and some of the cyclin-dependent kinases (CDKs). After a brief summary of the molecular and cell biology of AD and HD this review covers what is known about the role of these three groups of kinases in the brain and in the pathogenesis of the two neurodegenerative disorders. The potential of targeting GSK3, p38 MAPK and CDKS as effective therapeutics is also discussed as is a brief discussion on the utilization of recently developed drugs that simultaneously target two or all three of these groups of kinases. Multi-kinase inhibitors either by themselves or in combination with strategies currently being used such as immunotherapy or secretase inhibitors for AD and knockdown for HD could represent a more effective therapeutic approach for these fatal neurodegenerative diseases.
Collapse
|
37
|
Stepien BK, Vaid S, Huttner WB. Length of the Neurogenic Period-A Key Determinant for the Generation of Upper-Layer Neurons During Neocortex Development and Evolution. Front Cell Dev Biol 2021; 9:676911. [PMID: 34055808 PMCID: PMC8155536 DOI: 10.3389/fcell.2021.676911] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 04/20/2021] [Indexed: 11/17/2022] Open
Abstract
The neocortex, a six-layer neuronal brain structure that arose during the evolution of, and is unique to, mammals, is the seat of higher order brain functions responsible for human cognitive abilities. Despite its recent evolutionary origin, it shows a striking variability in size and folding complexity even among closely related mammalian species. In most mammals, cortical neurogenesis occurs prenatally, and its length correlates with the length of gestation. The evolutionary expansion of the neocortex, notably in human, is associated with an increase in the number of neurons, particularly within its upper layers. Various mechanisms have been proposed and investigated to explain the evolutionary enlargement of the human neocortex, focussing in particular on changes pertaining to neural progenitor types and their division modes, driven in part by the emergence of human-specific genes with novel functions. These led to an amplification of the progenitor pool size, which affects the rate and timing of neuron production. In addition, in early theoretical studies, another mechanism of neocortex expansion was proposed—the lengthening of the neurogenic period. A critical role of neurogenic period length in determining neocortical neuron number was subsequently supported by mathematical modeling studies. Recently, we have provided experimental evidence in rodents directly supporting the mechanism of extending neurogenesis to specifically increase the number of upper-layer cortical neurons. Moreover, our study examined the relationship between cortical neurogenesis and gestation, linking the extension of the neurogenic period to the maternal environment. As the exact nature of factors promoting neurogenic period prolongation, as well as the generalization of this mechanism for evolutionary distinct lineages, remain elusive, the directions for future studies are outlined and discussed.
Collapse
Affiliation(s)
- Barbara K Stepien
- Max Planck Institute of Molecular Cell Biology and Genetics, Max Planck Society (MPG), Munich, Germany.,Institute of Anatomy, Faculty of Medicine Carl Gustav Carus, School of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Max Planck Society (MPG), Munich, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Max Planck Society (MPG), Munich, Germany
| |
Collapse
|
38
|
Schörnig M, Taverna E. A Closer Look to the Evolution of Neurons in Humans and Apes Using Stem-Cell-Derived Model Systems. Front Cell Dev Biol 2021; 9:661113. [PMID: 33968936 PMCID: PMC8097028 DOI: 10.3389/fcell.2021.661113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
The cellular, molecular and functional comparison of neurons from closely related species is crucial in evolutionary neurobiology. The access to living tissue and post-mortem brains of humans and non-human primates is limited and the state of the tissue might not allow recapitulating important species-specific differences. A valid alternative is offered by neurons derived from induced pluripotent stem cells (iPSCs) obtained from humans and non-human apes and primates. We will review herein the contribution of iPSCs-derived neuronal models to the field of evolutionary neurobiology, focusing on species-specific aspects of neuron’s cell biology and timing of maturation. In addition, we will discuss the use of iPSCs for the study of ancient human traits.
Collapse
Affiliation(s)
- Maria Schörnig
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Elena Taverna
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| |
Collapse
|
39
|
Schnabl J, Litz MPH, Schneider C, PenkoffLidbeck N, Bashiruddin S, Schwartz MS, Alligood K, Devoto SH, Barresi MJF. Characterizing the diverse cells that associate with the developing commissures of the zebrafish forebrain. Dev Neurobiol 2021; 81:671-695. [PMID: 33314626 DOI: 10.1002/dneu.22801] [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: 07/17/2020] [Revised: 10/20/2020] [Accepted: 12/08/2020] [Indexed: 01/02/2023]
Abstract
During embryonic development of bilaterally symmetrical organisms, neurons send axons across the midline at specific points to connect the two halves of the nervous system with a commissure. Little is known about the cells at the midline that facilitate this tightly regulated process. We exploit the conserved process of vertebrate embryonic development in the zebrafish model system to elucidate the identity of cells at the midline that may facilitate postoptic (POC) and anterior commissure (AC) development. We have discovered that three different gfap+ astroglial cell morphologies persist in contact with pathfinding axons throughout commissure formation. Similarly, olig2+ progenitor cells occupy delineated portions of the postoptic and anterior commissures where they act as multipotent, neural progenitors. Moreover, we conclude that both gfap+ and olig2+ progenitor cells give rise to neuronal populations in both the telencephalon and diencephalon; however, these varied cell populations showed significant developmental timing differences between the telencephalon and diencephalon. Lastly, we also showed that fli1a+ mesenchymal cells migrate along the presumptive commissure regions before and during midline axon crossing. Furthermore, following commissure maturation, specific blood vessels formed at the midline of the POC and immediately ventral and parallel to the AC. This comprehensive account of the cellular populations that correlate with the timing and position of commissural axon pathfinding has supported the conceptual modeling and identification of the early forebrain architecture that may be necessary for proper commissure development.
Collapse
Affiliation(s)
- Jake Schnabl
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA
| | - Mackenzie P H Litz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caitlin Schneider
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,McGill University, Montreal, QC, Canada
| | | | - Sarah Bashiruddin
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Family Medicine Assoc, Westfield, MA, USA
| | - Morgan S Schwartz
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kristin Alligood
- Department of Biological Sciences, Smith College, Northampton, MA, USA.,Farmers Conservation Alliance, Hood River, OR, USA
| | | | - Michael J F Barresi
- Department of Molecular and Cellular Biology, University of Massachusetts, Amherst, MA, USA.,Department of Biological Sciences, Smith College, Northampton, MA, USA
| |
Collapse
|
40
|
Friedrich P, Forkel SJ, Amiez C, Balsters JH, Coulon O, Fan L, Goulas A, Hadj-Bouziane F, Hecht EE, Heuer K, Jiang T, Latzman RD, Liu X, Loh KK, Patil KR, Lopez-Persem A, Procyk E, Sallet J, Toro R, Vickery S, Weis S, Wilson CRE, Xu T, Zerbi V, Eickoff SB, Margulies DS, Mars RB, Thiebaut de Schotten M. Imaging evolution of the primate brain: the next frontier? Neuroimage 2021; 228:117685. [PMID: 33359344 PMCID: PMC7116589 DOI: 10.1016/j.neuroimage.2020.117685] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 11/22/2022] Open
Abstract
Evolution, as we currently understand it, strikes a delicate balance between animals' ancestral history and adaptations to their current niche. Similarities between species are generally considered inherited from a common ancestor whereas observed differences are considered as more recent evolution. Hence comparing species can provide insights into the evolutionary history. Comparative neuroimaging has recently emerged as a novel subdiscipline, which uses magnetic resonance imaging (MRI) to identify similarities and differences in brain structure and function across species. Whereas invasive histological and molecular techniques are superior in spatial resolution, they are laborious, post-mortem, and oftentimes limited to specific species. Neuroimaging, by comparison, has the advantages of being applicable across species and allows for fast, whole-brain, repeatable, and multi-modal measurements of the structure and function in living brains and post-mortem tissue. In this review, we summarise the current state of the art in comparative anatomy and function of the brain and gather together the main scientific questions to be explored in the future of the fascinating new field of brain evolution derived from comparative neuroimaging.
Collapse
Affiliation(s)
- Patrick Friedrich
- Brain Connectivity and Behaviour Laboratory, Sorbonne Universities, Paris, France; Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA, University of Bordeaux, Bordeaux, France; Institute of Neuroscience and Medicine (Brain & Behaviour, INM-7), Research Center Jülich, Germany.
| | - Stephanie J Forkel
- Brain Connectivity and Behaviour Laboratory, Sorbonne Universities, Paris, France; Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA, University of Bordeaux, Bordeaux, France; Centre for Neuroimaging Sciences, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Céline Amiez
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France
| | - Joshua H Balsters
- Department of Psychology, Royal Holloway University of London, United Kingdom
| | - Olivier Coulon
- Institut de Neurosciences de la Timone, Aix Marseille Univ, CNRS, UMR 7289, Marseille, France; Institute for Language, Communication, and the Brain, Aix-Marseille University, Marseille, France
| | - Lingzhong Fan
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Alexandros Goulas
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg University, Hamburg, Germany
| | - Fadila Hadj-Bouziane
- Lyon Neuroscience Research Center, ImpAct Team, INSERM U1028, CNRS UMR5292, Université Lyon 1, Bron, France
| | - Erin E Hecht
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, United States
| | - Katja Heuer
- Center for Research and Interdisciplinarity (CRI), Université de Paris, Inserm, Paris 75004, France; Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Tianzi Jiang
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; The Queensland Brain Institute, University of Queensland, Brisbane QLD 4072, Australia
| | - Robert D Latzman
- Department of Psychology, Georgia State University, Atlanta, United States
| | - Xiaojin Liu
- Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; Institute of Neuroscience and Medicine (Brain & Behaviour, INM-7), Research Center Jülich, Germany
| | - Kep Kee Loh
- Institut de Neurosciences de la Timone, Aix Marseille Univ, CNRS, UMR 7289, Marseille, France; Institute for Language, Communication, and the Brain, Aix-Marseille University, Marseille, France
| | - Kaustubh R Patil
- Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; Institute of Neuroscience and Medicine (Brain & Behaviour, INM-7), Research Center Jülich, Germany
| | - Alizée Lopez-Persem
- Frontlab, Institut du Cerveau et de la Moelle épinière (ICM), UPMC UMRS 1127, Inserm U 1127, CNRS UMR 7225, Paris, France; Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Emmanuel Procyk
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France
| | - Jerome Sallet
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France; Wellcome Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford, United Kingdom
| | - Roberto Toro
- Center for Research and Interdisciplinarity (CRI), Université de Paris, Inserm, Paris 75004, France; Neuroscience department, Institut Pasteur, UMR 3571, CNRS, Université de Paris, Paris 75015, France
| | - Sam Vickery
- Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; Institute of Neuroscience and Medicine (Brain & Behaviour, INM-7), Research Center Jülich, Germany
| | - Susanne Weis
- Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; Institute of Neuroscience and Medicine (Brain & Behaviour, INM-7), Research Center Jülich, Germany
| | - Charles R E Wilson
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute, U1208 Bron, France
| | - Ting Xu
- Child Mind Institute, New York, United States
| | - Valerio Zerbi
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Simon B Eickoff
- Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; Institute of Neuroscience and Medicine (Brain & Behaviour, INM-7), Research Center Jülich, Germany
| | - Daniel S Margulies
- Brain Connectivity and Behaviour Laboratory, Sorbonne Universities, Paris, France; Integrative Neuroscience and Cognition Center (UMR 8002), Centre National de la Recherche Scientifique (CNRS) and Université de Paris, 75006, Paris, France
| | - Rogier B Mars
- Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behaviour Laboratory, Sorbonne Universities, Paris, France; Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
41
|
Jiang T, Hu S, Dai S, Yi Y, Wang T, Li X, Luo M, Li K, Chen L, Wang H, Xu D. Programming changes of hippocampal miR-134-5p/SOX2 signal mediate the susceptibility to depression in prenatal dexamethasone-exposed female offspring. Cell Biol Toxicol 2021; 38:69-86. [PMID: 33619658 DOI: 10.1007/s10565-021-09590-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/09/2021] [Indexed: 12/21/2022]
Abstract
Depression is a neuropsychiatric disorder and has intrauterine developmental origins. This study aimed to confirm the depression susceptibility in offspring rats induced by prenatal dexamethasone exposure (PDE) and to further explore the intrauterine programming mechanism. Wistar rats were injected with dexamethasone (0.2 mg/kg·d) subcutaneously during the gestational days 9-20 and part of the offspring was given chronic stress at postnatal weeks 10-12. Behavioral results showed that the adult PDE female offspring was susceptible to depression, accompanied by increased hippocampal miR-134-5p expression and decreased sex-determining region Y-box 2 (SOX2) expression, as well as disorders of neural progenitor cells proliferation and hippocampal neurogenesis. The PDE female fetal rats presented consistent changes with the adult offspring, accompanied by the upregulation of glucocorticoid receptor (GR) expression and decreased sirtuin 1 (SIRT1) expression. We further found that the H3K9ac level of the miR-134-5p promoter was significantly increased in the PDE fetal hippocampus, as well as in adult offspring before and after chronic stress. In vitro, the changes of GR/SIRT1/miR-134-5p/SOX2 signal by dexamethasone were consistent with in vivo experiments, which could be reversed by GR receptor antagonist, SIRT1 agonist, and miR-134-5p inhibitor. This study confirmed that PDE led to an increased expression level as well as H3K9ac level of miR-134-5p by activating the GR/SIRT1 pathway in the fetal hippocampus and then inhibited the SOX2 expression. The programming effect mediated by the abnormal epigenetic modification could last from intrauterine to adulthood, which constitutes the intrauterine programming mechanism leading to hippocampal neurogenesis disorders and depression susceptibility in female offspring. Intrauterine programming mechanism for the increased depressive susceptibility in adult female offspring by prenatal dexamethasone exposure (PDE). GR, glucocorticoid receptor; SIRT1, sirtuin 1; SOX2, sex-determining region Y-box 2; NPCs, neuroprogenitor cells; H3K9ac, histone 3 lysine 9 acetylation; GRE, glucocorticoid response element.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Shuwei Hu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Shiyun Dai
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Yiwen Yi
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Tingting Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Xufeng Li
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Mingcui Luo
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China
| | - Ke Li
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University, Wuhan, 430071, China
| | - Liaobin Chen
- Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China
| | - Dan Xu
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan, 430071, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| |
Collapse
|
42
|
Hernández-Núñez I, Robledo D, Mayeur H, Mazan S, Sánchez L, Adrio F, Barreiro-Iglesias A, Candal E. Loss of Active Neurogenesis in the Adult Shark Retina. Front Cell Dev Biol 2021; 9:628721. [PMID: 33644067 PMCID: PMC7905061 DOI: 10.3389/fcell.2021.628721] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/12/2021] [Indexed: 01/09/2023] Open
Abstract
Neurogenesis is the process by which progenitor cells generate new neurons. As development progresses neurogenesis becomes restricted to discrete neurogenic niches, where it persists during postnatal life. The retina of teleost fishes is thought to proliferate and produce new cells throughout life. Whether this capacity may be an ancestral characteristic of gnathostome vertebrates is completely unknown. Cartilaginous fishes occupy a key phylogenetic position to infer ancestral states fixed prior to the gnathostome radiation. Previous work from our group revealed that the juvenile retina of the catshark Scyliorhinus canicula, a cartilaginous fish, shows active proliferation and neurogenesis. Here, we compared the morphology and proliferative status of the retina in catshark juveniles and adults. Histological and immunohistochemical analyses revealed an important reduction in the size of the peripheral retina (where progenitor cells are mainly located), a decrease in the thickness of the inner nuclear layer (INL), an increase in the thickness of the inner plexiform layer and a decrease in the cell density in the INL and in the ganglion cell layer in adults. Contrary to what has been reported in teleost fish, mitotic activity in the catshark retina was virtually absent after sexual maturation. Based on these results, we carried out RNA-Sequencing (RNA-Seq) analyses comparing the retinal transcriptome of juveniles and adults, which revealed a statistically significant decrease in the expression of many genes involved in cell proliferation and neurogenesis in adult catsharks. Our RNA-Seq data provides an excellent resource to identify new signaling pathways controlling neurogenesis in the vertebrate retina.
Collapse
Affiliation(s)
- Ismael Hernández-Núñez
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Hélène Mayeur
- CNRS, Sorbonne Universités, UPMC Univ Paris 06, UMR7232, Observatoire Océanologique, Banyuls-sur-mer, France
| | - Sylvie Mazan
- CNRS, Sorbonne Universités, UPMC Univ Paris 06, UMR7232, Observatoire Océanologique, Banyuls-sur-mer, France
| | - Laura Sánchez
- Departamento de Zooloxía, Xenética e Antropoloxía Física, Facultade de Veterinaria, Universidade de Santiago de Compostela, Lugo, Spain
| | - Fátima Adrio
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Antón Barreiro-Iglesias
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Eva Candal
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
43
|
Chen R, Hou Y, Connell M, Zhu S. Homeodomain protein Six4 prevents the generation of supernumerary Drosophila type II neuroblasts and premature differentiation of intermediate neural progenitors. PLoS Genet 2021; 17:e1009371. [PMID: 33556050 PMCID: PMC7895384 DOI: 10.1371/journal.pgen.1009371] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 02/19/2021] [Accepted: 01/15/2021] [Indexed: 11/19/2022] Open
Abstract
In order to boost the number and diversity of neurons generated from neural stem cells, intermediate neural progenitors (INPs) need to maintain their homeostasis by avoiding both dedifferentiation and premature differentiation. Elucidating how INPs maintain homeostasis is critical for understanding the generation of brain complexity and various neurological diseases resulting from defects in INP development. Here we report that Six4 expressed in Drosophila type II neuroblast (NB) lineages prevents the generation of supernumerary type II NBs and premature differentiation of INPs. We show that loss of Six4 leads to supernumerary type II NBs likely due to dedifferentiation of immature INPs (imINPs). We provide data to further demonstrate that Six4 inhibits the expression and activity of PntP1 in imINPs in part by forming a trimeric complex with Earmuff and PntP1. Furthermore, knockdown of Six4 exacerbates the loss of INPs resulting from the loss of PntP1 by enhancing ectopic Prospero expression in imINPs, suggesting that Six4 is also required for preventing premature differentiation of INPs. Taken together, our work identified a novel transcription factor that likely plays important roles in maintaining INP homeostasis. Intermediate neural progenitors (INPs) are descendants of neural stem cells that can proliferate for a short term to amplify the number of nerve cells generated in the brain. INPs play critical roles in determining how big and complex a brain can grow. To perform their function, INPs need to maintain their own population and must not adopt the identity of neural stem cells, a process called dedifferentiation, or acquire the fate of their own daughter cells and stop proliferation too soon, a process called premature differentiation. However, how INPs avoid dedifferentiation and premature differentiation is not fully understood. In this study, we identified a protein called Six4 as a novel factor that plays important roles in preventing the generation of extra neural stem cells and premature differentiation of INPs in developing fruit fly brains. We described how Six4 functionally and physically interacts with other factors that are involved in regulating INP cell fate specification. Our work provides novel insights into the mechanisms regulating INP development and could have important implications in understanding how complex brains are generated during normal development and how abnormal brain development or brain tumor can occur when INPs fail to avoid premature differentiation or dedifferentiation.
Collapse
Affiliation(s)
- Rui Chen
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Yanjun Hou
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Marisa Connell
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, United States of America
| | - Sijun Zhu
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, New York, United States of America
- * E-mail:
| |
Collapse
|
44
|
Exner CRT, Willsey HR. Xenopus leads the way: Frogs as a pioneering model to understand the human brain. Genesis 2021; 59:e23405. [PMID: 33369095 PMCID: PMC8130472 DOI: 10.1002/dvg.23405] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/12/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
From its long history in the field of embryology to its recent advances in genetics, Xenopus has been an indispensable model for understanding the human brain. Foundational studies that gave us our first insights into major embryonic patterning events serve as a crucial backdrop for newer avenues of investigation into organogenesis and organ function. The vast array of tools available in Xenopus laevis and Xenopus tropicalis allows interrogation of developmental phenomena at all levels, from the molecular to the behavioral, and the application of CRISPR technology has enabled the investigation of human disorder risk genes in a higher-throughput manner. As the only major tetrapod model in which all developmental stages are easily manipulated and observed, frogs provide the unique opportunity to study organ development from the earliest stages. All of these features make Xenopus a premier model for studying the development of the brain, a notoriously complex process that demands an understanding of all stages from fertilization to organogenesis and beyond. Importantly, core processes of brain development are conserved between Xenopus and human, underlining the advantages of this model. This review begins by summarizing discoveries made in amphibians that form the cornerstones of vertebrate neurodevelopmental biology and goes on to discuss recent advances that have catapulted our understanding of brain development in Xenopus and in relation to human development and disease. As we engage in a new era of patient-driven gene discovery, Xenopus offers exceptional potential to uncover conserved biology underlying human brain disorders and move towards rational drug design.
Collapse
Affiliation(s)
- Cameron R T Exner
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, 94143, USA
| | - Helen Rankin Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California, 94143, USA
| |
Collapse
|
45
|
de Agustín-Durán D, Mateos-White I, Fabra-Beser J, Gil-Sanz C. Stick around: Cell-Cell Adhesion Molecules during Neocortical Development. Cells 2021; 10:118. [PMID: 33435191 PMCID: PMC7826847 DOI: 10.3390/cells10010118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/29/2020] [Accepted: 01/07/2021] [Indexed: 12/21/2022] Open
Abstract
The neocortex is an exquisitely organized structure achieved through complex cellular processes from the generation of neural cells to their integration into cortical circuits after complex migration processes. During this long journey, neural cells need to establish and release adhesive interactions through cell surface receptors known as cell adhesion molecules (CAMs). Several types of CAMs have been described regulating different aspects of neurodevelopment. Whereas some of them mediate interactions with the extracellular matrix, others allow contact with additional cells. In this review, we will focus on the role of two important families of cell-cell adhesion molecules (C-CAMs), classical cadherins and nectins, as well as in their effectors, in the control of fundamental processes related with corticogenesis, with special attention in the cooperative actions among the two families of C-CAMs.
Collapse
Affiliation(s)
| | | | | | - Cristina Gil-Sanz
- Neural Development Laboratory, Instituto Universitario de Biomedicina y Biotecnología (BIOTECMED) and Departamento de Biología Celular, Facultat de Biología, Universidad de Valencia, 46100 Burjassot, Spain; (D.d.A.-D.); (I.M.-W.); (J.F.-B.)
| |
Collapse
|
46
|
Bourin M. Neurogenesis and Neuroplasticity in Major Depression: Its Therapeutic Implication. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:157-173. [PMID: 33834400 DOI: 10.1007/978-981-33-6044-0_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The neurochemical model of depression, based on monoaminergic theories, does not allow on its own to understand the mechanism of action of antidepressants. This approach does not explain the gap between the immediate biochemical modulations induced by antidepressants and the time required for their clinical action. Several hypotheses have been developed to try to explain more precisely the action of these molecules, each of them involving mechanisms of receptor regulation. At the same time, data on the neuroanatomy of depression converge toward the existence of specific lesions of this pathology. This chapter aims to provide an overview of recent advances in understanding the mechanisms of neural plasticity involved in pathophysiology depression and in its treatment.
Collapse
Affiliation(s)
- Michel Bourin
- Neurobiology of Mood Disorders, University of Nantes, Nantes, France.
| |
Collapse
|
47
|
Chowdhury S, Ghosh S. Next Generation Sequencing and Stem Cells. Stem Cells 2021. [DOI: 10.1007/978-981-16-1638-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
48
|
Gonda Y, Namba T, Hanashima C. Beyond Axon Guidance: Roles of Slit-Robo Signaling in Neocortical Formation. Front Cell Dev Biol 2020; 8:607415. [PMID: 33425915 PMCID: PMC7785817 DOI: 10.3389/fcell.2020.607415] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
The formation of the neocortex relies on intracellular and extracellular signaling molecules that are involved in the sequential steps of corticogenesis, ranging from the proliferation and differentiation of neural progenitor cells to the migration and dendrite formation of neocortical neurons. Abnormalities in these steps lead to disruption of the cortical structure and circuit, and underly various neurodevelopmental diseases, including dyslexia and autism spectrum disorder (ASD). In this review, we focus on the axon guidance signaling Slit-Robo, and address the multifaceted roles of Slit-Robo signaling in neocortical development. Recent studies have clarified the roles of Slit-Robo signaling not only in axon guidance but also in progenitor cell proliferation and migration, and the maturation of neocortical neurons. We further discuss the etiology of neurodevelopmental diseases, which are caused by defects in Slit-Robo signaling during neocortical formation.
Collapse
Affiliation(s)
- Yuko Gonda
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Neuroscience Center, HiLIFE – Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Carina Hanashima
- Faculty of Education and Integrated Arts and Sciences, Waseda University, Tokyo, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
49
|
Abstract
Neural stem cells (NSCs) persist into adulthood in the subgranular zone (SGZ) of the dentate gyrus in the hippocampus and in the ventricular-subventricular zone (V-SVZ) of the lateral ventricles, where they generate new neurons and glia cells that contribute to neural plasticity. A better understanding of the developmental process that enables NSCs to persist beyond development will provide insight into factors that determine the size and properties of the adult NSC pool and thus the capacity for life-long neurogenesis in the adult mammalian brain. We review current knowledge regarding the developmental origins of adult NSCs and the developmental process by which embryonic NSCs transition into their adult form. We also discuss potential mechanisms that might regulate proper establishment of the adult NSC pool, and propose future directions of research that will be key to unraveling how NSCs transform to establish the adult NSC pool in the mammalian brain.
Collapse
Affiliation(s)
- Allison M Bond
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, United States; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
50
|
Namba T, Nardelli J, Gressens P, Huttner WB. Metabolic Regulation of Neocortical Expansion in Development and Evolution. Neuron 2020; 109:408-419. [PMID: 33306962 DOI: 10.1016/j.neuron.2020.11.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/19/2020] [Accepted: 11/13/2020] [Indexed: 12/18/2022]
Abstract
The neocortex, the seat of our higher cognitive abilities, has expanded in size during the evolution of certain mammals such as primates, including humans. This expansion occurs during development and is linked to the proliferative capacity of neural stem and progenitor cells (NPCs) in the neocortex. A number of cell-intrinsic and cell-extrinsic factors have been implicated in increasing NPC proliferative capacity. However, NPC metabolism has only recently emerged as major regulator of NPC proliferation. In this Perspective, we summarize recent insights into the role of NPC metabolism in neocortical development and neurodevelopmental disorders and its relevance for neocortex evolution. We discuss certain human-specific genes and microcephaly-implicated genes that operate in, or at, the mitochondria of NPCs and stimulate their proliferation by promoting glutaminolysis. We also discuss other metabolic pathways and develop a perspective on how metabolism mechanistically regulates NPC proliferation in neocortical development and how this contributed to neocortex evolution.
Collapse
Affiliation(s)
- Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany; Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | | | - Pierre Gressens
- Université de Paris, NeuroDiderot, Inserm, 75019 Paris, France.
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|