1
|
Imamura M, Yoshino M, Kawasaki H. Investigation of the development and evolution of the mammalian cerebrum using gyrencephalic ferrets. Eur J Cell Biol 2024; 103:151466. [PMID: 39546916 DOI: 10.1016/j.ejcb.2024.151466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Mammalian brains have evolved a neocortex, which has diverged in size and morphology in different species over the course of evolution. In some mammals, a substantial increase in the number of neurons and glial cells resulted in the expansion and folding of the cerebrum, and it is believed that these evolutionary changes contributed to the acquisition of higher cognitive abilities in mammals. However, their underlying molecular and cellular mechanisms remain insufficiently elucidated. A major difficulty in addressing these mechanisms stemmed from the lack of appropriate animal models, as conventional experimental animals such as mice and rats have small brains without structurally obvious folds. Therefore, researchers including us have focused on using ferrets instead of mice and rats. Ferrets are domesticated carnivorous mammals with a gyrencephalic cerebrum, and, notably, they are amenable to genetic manipulations including in utero electroporation to knock out genes in the cerebrum. In this review, we highlight recent research into the mechanisms underlying the development and evolution of cortical folds using ferrets.
Collapse
Affiliation(s)
- Masanori Imamura
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Mayuko Yoshino
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
2
|
Bury LAD, Fu S, Wynshaw-Boris A. Neuronal lineage tracing from progenitors in human cortical organoids reveals mechanisms of neuronal production, diversity, and disease. Cell Rep 2024; 43:114862. [PMID: 39395167 DOI: 10.1016/j.celrep.2024.114862] [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: 08/20/2023] [Revised: 08/14/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024] Open
Abstract
The contribution of progenitor subtypes to generating the billions of neurons produced during human cortical neurogenesis is not well understood. We developed the cortical organoid lineage-tracing (COR-LT) system for human cortical organoids. Differential fluorescent reporter activation in distinct progenitor cells leads to permanent reporter expression, enabling the progenitor cell lineage of neurons to be determined. Surprisingly, nearly all excitatory neurons produced in cortical organoids were generated indirectly from intermediate progenitor cells. Additionally, neurons of different progenitor lineages were transcriptionally distinct. Isogenic lines made from an autistic individual with and without a likely pathogenic CTNNB1 variant demonstrated that the variant substantially altered the proportion of neurons derived from specific progenitor cell lineages, as well as the lineage-specific transcriptional profiles of these neurons, suggesting a pathogenic mechanism for this mutation. These results suggest individual progenitor subtypes play roles in generating the diverse neurons of the human cerebral cortex.
Collapse
Affiliation(s)
- Luke A D Bury
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| | - Shuai Fu
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Cleveland, OH 44195, USA
| | - Anthony Wynshaw-Boris
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA.
| |
Collapse
|
3
|
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
|
4
|
Mao Y, Suryawanshi A, Patial S, Saini Y. Airspaces-derived exosomes contain disease-relevant protein signatures in a mouse model of cystic fibrosis (CF)-like mucoinflammatory lung disease. Front Pharmacol 2024; 15:1460692. [PMID: 39386033 PMCID: PMC11461968 DOI: 10.3389/fphar.2024.1460692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Exosomes, membrane-bound extracellular vesicles, ranging from approximately 30-200 nm in diameter, are released by almost all cell types and play critical roles in intercellular communication. In response to the prevailing stress, the exosome-bound protein signatures vary in abundance and composition. To identify the bronchoalveolar lavage fluid (BALF) exosome-bound proteins associated with mucoinflammatory lung disease and to gain insights into their functional implications, we compared BALF exosomes-derived proteins from adult Scnn1b transgenic (Scnn1b-Tg+) and wild type (WT) mice. A total of 3,144 and 3,119 proteins were identified in BALF exosomes from Scnn1b-Tg+ and WT mice, respectively. Using cutoff criteria (Log2 fold-change > 1 and adjusted p-value < 0.05), the comparison of identified proteins revealed 127 and 30 proteins that were significantly upregulated and downregulated, respectively, in Scnn1b-Tg+ versus WT mice. In addition, 52 and 27 proteins were exclusively enriched in Scnn1b-Tg+ and WT mice, respectively. The identified exosome-bound proteins from the homeostatic airspaces of WT mice were mostly relevant to the normal physiological processes. The protein signatures enriched in the BALF exosomes of Scnn1b-Tg+ mice were relevant to macrophage activation and mucoinflammatory processes. Ingenuity pathway analyses revealed that the enriched proteins in Scnn1b-Tg+ mice contributed to the inflammatory responses and antimicrobial defense pathways. Selective proteins including, RETNLA/FIZZ1, LGALS3/Galectin-3, S100A8/MRP8, and CHIL3/YM1 were immunolocalized to specific cell types. The comparative analysis between enriched BALF exosome proteins and previously identified differentially upregulated genes in Scnn1b-Tg+ versus WT mice suggested that the compartment-/cell-specific upregulation in gene expression dictates the enrichment of their respective proteins in the lung airspaces. Taken together, this study demonstrates that the BALF exosome-bound protein signatures reflect disease-relevant disturbances. Our findings suggest that the exosomes carry disease-relevant protein signatures that can be used as a diagnostic as well as predictive biomarkers for mucoinflammatory lung disease.
Collapse
Affiliation(s)
- Yun Mao
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sonika Patial
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, United States
| | - Yogesh Saini
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Deng H, Tong S, Shen D, Zhang S, Fu Y. The characteristics of excitatory lineage differentiation and the developmental conservation in Reeler neocortex. Cell Prolif 2024; 57:e13587. [PMID: 38084819 PMCID: PMC11056708 DOI: 10.1111/cpr.13587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/21/2023] [Accepted: 11/29/2023] [Indexed: 04/30/2024] Open
Abstract
The majority of neocortical projection neurons are generated indirectly from radial glial cells (RGCs) mediated by intermediate progenitor cells (IPCs) in mice. IPCs are thought to be a great breakthrough in the evolutionary expansion of the mammalian neocortex. However, the precise ratio of neuron production from IPCs and characteristics of RGC differentiation process are still unclear. Our study revealed that direct neurogenesis was seldom observed and increased slightly at late embryonic stage. Besides, we conducted retrovirus sparse labelling combined carboxyfluorescein diacetate succinimide ester (CFSE) and Tbr2-CreER strain to reconstruct individual lineage tree in situ. The lineage trees simulated the output of RGCs at per round of division in sequence with high temporal, spatial and cellular resolution at P7. We then demonstrated that only 1.90% of neurons emanated from RGCs directly in mouse cerebral neocortex and 79.33% of RGCs contributed to the whole clones through IPCs. The contribution of indirect neurogenesis was underestimated previously because approximately a quarter of IPC-derived neurons underwent apoptosis. Here, we also showed that abundant IPCs from first-generation underwent self-renewing division and generated four neurons ultimately. We confirmed that the intermediate proliferative progenitors expressed higher Cux2 characteristically at early embryonic stage. Finally, we validated that the characteristics of neurogenetic process in lineages and developmental fate of neurons were conserved in Reeler mice. This study contributes to further understanding of neurogenesis in neocortical development.
Collapse
Affiliation(s)
- Huan‐Huan Deng
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Shi‐Yuan Tong
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Dan Shen
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Shu‐Qing Zhang
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yinghui Fu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| |
Collapse
|
8
|
Coquand L, Brunet Avalos C, Macé AS, Farcy S, Di Cicco A, Lampic M, Wimmer R, Bessières B, Attie-Bitach T, Fraisier V, Sens P, Guimiot F, Brault JB, Baffet AD. A cell fate decision map reveals abundant direct neurogenesis bypassing intermediate progenitors in the human developing neocortex. Nat Cell Biol 2024; 26:698-709. [PMID: 38548890 PMCID: PMC11098750 DOI: 10.1038/s41556-024-01393-z] [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: 09/25/2022] [Accepted: 02/29/2024] [Indexed: 05/03/2024]
Abstract
The human neocortex has undergone strong evolutionary expansion, largely due to an increased progenitor population, the basal radial glial cells. These cells are responsible for the production of a diversity of cell types, but the successive cell fate decisions taken by individual progenitors remain unknown. Here we developed a semi-automated live/fixed correlative imaging method to map basal radial glial cell division modes in early fetal tissue and cerebral organoids. Through the live analysis of hundreds of dividing progenitors, we show that basal radial glial cells undergo abundant symmetric amplifying divisions, and frequent self-consuming direct neurogenic divisions, bypassing intermediate progenitors. These direct neurogenic divisions are more abundant in the upper part of the subventricular zone. We furthermore demonstrate asymmetric Notch activation in the self-renewing daughter cells, independently of basal fibre inheritance. Our results reveal a remarkable conservation of fate decisions in cerebral organoids, supporting their value as models of early human neurogenesis.
Collapse
Affiliation(s)
- Laure Coquand
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
- Sorbonne Université, Ecole Doctorale complexité du vivant, Paris, France
| | | | - Anne-Sophie Macé
- UMR 144-Cell and Tissue Imaging Facility (PICT-IBiSA), CNRS-Institut Curie, Paris, France
| | - Sarah Farcy
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | | | - Marusa Lampic
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Ryszard Wimmer
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
- Sorbonne Université, Ecole Doctorale complexité du vivant, Paris, France
| | - Betina Bessières
- UF Embryofœtopathologie, Hopital Necker-enfants malades, Paris, France
| | | | - Vincent Fraisier
- UMR 144-Cell and Tissue Imaging Facility (PICT-IBiSA), CNRS-Institut Curie, Paris, France
| | - Pierre Sens
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - Fabien Guimiot
- UF de Fœtopathologie - Université de Paris et Inserm UMR1141, Hôpital Robert Debré, Paris, France
| | | | - Alexandre D Baffet
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France.
- Institut national de la santé et de la recherche médicale, Paris, France.
| |
Collapse
|
9
|
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
|
10
|
Stepien BK, Wielockx B. From Vessels to Neurons-The Role of Hypoxia Pathway Proteins in Embryonic Neurogenesis. Cells 2024; 13:621. [PMID: 38607059 PMCID: PMC11012138 DOI: 10.3390/cells13070621] [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: 02/28/2024] [Revised: 03/20/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Embryonic neurogenesis can be defined as a period of prenatal development during which divisions of neural stem and progenitor cells give rise to neurons. In the central nervous system of most mammals, including humans, the majority of neocortical neurogenesis occurs before birth. It is a highly spatiotemporally organized process whose perturbations lead to cortical malformations and dysfunctions underlying neurological and psychiatric pathologies, and in which oxygen availability plays a critical role. In case of deprived oxygen conditions, known as hypoxia, the hypoxia-inducible factor (HIF) signaling pathway is activated, resulting in the selective expression of a group of genes that regulate homeostatic adaptations, including cell differentiation and survival, metabolism and angiogenesis. While a physiological degree of hypoxia is essential for proper brain development, imbalanced oxygen levels can adversely affect this process, as observed in common obstetrical pathologies such as prematurity. This review comprehensively explores and discusses the current body of knowledge regarding the role of hypoxia and the HIF pathway in embryonic neurogenesis of the mammalian cortex. Additionally, it highlights existing gaps in our understanding, presents unanswered questions, and provides avenues for future research.
Collapse
Affiliation(s)
- Barbara K. Stepien
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Experimental Centre, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
11
|
Sokpor G, Kerimoglu C, Ulmke PA, Pham L, Nguyen HD, Brand-Saberi B, Staiger JF, Fischer A, Nguyen HP, Tuoc T. H3 Acetylation-Induced Basal Progenitor Generation and Neocortex Expansion Depends on the Transcription Factor Pax6. BIOLOGY 2024; 13:68. [PMID: 38392287 PMCID: PMC10886678 DOI: 10.3390/biology13020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/24/2024]
Abstract
Enrichment of basal progenitors (BPs) in the developing neocortex is a central driver of cortical enlargement. The transcription factor Pax6 is known as an essential regulator in generation of BPs. H3 lysine 9 acetylation (H3K9ac) has emerged as a crucial epigenetic mechanism that activates the gene expression program required for BP pool amplification. In this current work, we applied immunohistochemistry, RNA sequencing, chromatin immunoprecipitation and sequencing, and the yeast two-hybrid assay to reveal that the BP-genic effect of H3 acetylation is dependent on Pax6 functionality in the developing mouse cortex. In the presence of Pax6, increased H3 acetylation caused BP pool expansion, leading to enhanced neurogenesis, which evoked expansion and quasi-convolution of the mouse neocortex. Interestingly, H3 acetylation activation exacerbates the BP depletion and corticogenesis reduction effect of Pax6 ablation in cortex-specific Pax6 mutants. Furthermore, we found that H3K9 acetyltransferase KAT2A/GCN5 interacts with Pax6 and potentiates Pax6-dependent transcriptional activity. This explains a genome-wide lack of H3K9ac, especially in the promoter regions of BP-genic genes, in the Pax6 mutant cortex. Together, these findings reveal a mechanistic coupling of H3 acetylation and Pax6 in orchestrating BP production and cortical expansion through the promotion of a BP gene expression program during cortical development.
Collapse
Affiliation(s)
- Godwin Sokpor
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
- Lincoln Medical School, University of Lincoln, Lincoln LN6 7TS, UK
| | - Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | | | - Linh Pham
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Hoang Duy Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Tran Tuoc
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| |
Collapse
|
12
|
Holst CB, Brøchner CB, Vitting‐Seerup K, Møllgård K. The HOPX and BLBP landscape and gliogenic regions in developing human brain. J Anat 2023; 243:23-38. [PMID: 36794762 PMCID: PMC10273337 DOI: 10.1111/joa.13844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Outer radial glial cells (oRGs) give rise to neurons and glial cells and contribute to cell migration and expansion in developing neocortex. HOPX has been described as a marker of oRGs and possible actor in glioblastomas. Recent years' evidence points to spatiotemporal differences in brain development which may have implications for the classification of cell types in the central nervous system and understanding of a range of neurological diseases. Using the Human Embryonic/Fetal Biobank, Institute of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark, HOPX and BLBP immunoexpression was investigated in developing frontal, parietal, temporal and occipital human neocortex, other cortical areas and brain stem regions to interrogate oRG and HOPX regional heterogeneity. Furthermore, usage of high-plex spatial profiling (Nanostring GeoMx® DSP) was tested on the same material. HOPX marked oRGs in several human developing brain regions as well as cells in known gliogenic areas but did not completely overlap with BLBP or GFAP. Interestingly, limbic structures (e.g. olfactory bulb, indusium griseum, entorhinal cortex, fimbria) showed more intense HOPX immunoreactivity than adjacent neocortex and in cerebellum and brain stem, HOPX and BLBP seemed to stain different cell populations in cerebellar cortex and corpus pontobulbare. DSP screening of corresponding regions indicated differences in cell type composition, vessel density and presence of apolipoproteins within and across regions and thereby confirming the importance of acknowledging time and place in developmental neuroscience.
Collapse
Affiliation(s)
- Camilla Bjørnbak Holst
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- DCCC Brain Tumor CenterCopenhagen University HospitalCopenhagenDenmark
| | - Christian Beltoft Brøchner
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Pathology, Center of Diagnostic InvestigationCopenhagen University HospitalCopenhagenDenmark
| | | | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
13
|
Mann B, Crawford JC, Reddy K, Lott J, Youn YH, Gao G, Guy C, Chou CH, Darnell D, Trivedi S, Bomme P, Loughran AJ, Thomas PG, Han YG, Tuomanen EI. Bacterial TLR2/6 Ligands Block Ciliogenesis, Derepress Hedgehog Signaling, and Expand the Neocortex. mBio 2023; 14:e0051023. [PMID: 37052506 PMCID: PMC10294647 DOI: 10.1128/mbio.00510-23] [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: 02/27/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Microbial components have a range of direct effects on the fetal brain. However, little is known about the cellular targets and molecular mechanisms that mediate these effects. Neural progenitor cells (NPCs) control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. We identify ventricular radial glia (vRG), the primary NPC, as the target of bacterial cell wall (BCW) generated during the antibiotic treatment of maternal pneumonia. BCW enhanced proliferative potential of vRGs by shortening the cell cycle and increasing self-renewal. Expanded vRGs propagated to increase neuronal output in all cortical layers. Remarkably, Toll-like receptor 2 (TLR2), which recognizes BCW, localized at the base of primary cilia in vRGs and the BCW-TLR2 interaction suppressed ciliogenesis leading to derepression of Hedgehog (HH) signaling and expansion of vRGs. We also show that TLR6 is an essential partner of TLR2 in this process. Surprisingly, TLR6 alone was required to set the number of cortical neurons under healthy conditions. These findings suggest that an endogenous signal from TLRs suppresses cortical expansion during normal development of the neocortex and that BCW antagonizes that signal through the TLR2/cilia/HH signaling axis changing brain structure and function. IMPORTANCE Fetal brain development in early gestation can be impacted by transplacental infection, altered metabolites from the maternal microbiome, or maternal immune activation. It is less well understood how maternal microbial subcomponents that cross the placenta, such as bacterial cell wall (BCW), directly interact with fetal neural progenitors and neurons and affect development. This scenario plays out in the clinic when BCW debris released during antibiotic therapy of maternal infection traffics to the fetal brain. This study identifies the direct interaction of BCW with TLR2/6 present on the primary cilium, the signaling hub on fetal neural progenitor cells (NPCs). NPCs control the size and architecture of the brain and understanding the mechanisms regulating NPCs is crucial to understanding brain developmental disorders. Within a window of vulnerability before the appearance of fetal immune cells, the BCW-TLR2/6 interaction results in the inhibition of ciliogenesis, derepression of Sonic Hedgehog signaling, excess proliferation of neural progenitors, and abnormal cortical architecture. In the first example of TLR signaling linked to Sonic Hedgehog, BCW/TLR2/6 appears to act during fetal brain morphogenesis to play a role in setting the total cell number in the neocortex.
Collapse
Affiliation(s)
- Beth Mann
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kavya Reddy
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Josi Lott
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yong Ha Youn
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Geli Gao
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ching-Heng Chou
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Daniel Darnell
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sanchit Trivedi
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Perrine Bomme
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Allister J. Loughran
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Young-Goo Han
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Elaine I. Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Andrews MG, Subramanian L, Salma J, Kriegstein AR. How mechanisms of stem cell polarity shape the human cerebral cortex. Nat Rev Neurosci 2022; 23:711-724. [PMID: 36180551 PMCID: PMC10571506 DOI: 10.1038/s41583-022-00631-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 11/09/2022]
Abstract
Apical-basal progenitor cell polarity establishes key features of the radial and laminar architecture of the developing human cortex. The unique diversity of cortical stem cell populations and an expansion of progenitor population size in the human cortex have been mirrored by an increase in the complexity of cellular processes that regulate stem cell morphology and behaviour, including their polarity. The study of human cells in primary tissue samples and human stem cell-derived model systems (such as cortical organoids) has provided insight into these processes, revealing that protein complexes regulate progenitor polarity by controlling cell membrane adherence within appropriate cortical niches and are themselves regulated by cytoskeletal proteins, signalling molecules and receptors, and cellular organelles. Studies exploring how cortical stem cell polarity is established and maintained are key for understanding the features of human brain development and have implications for neurological dysfunction.
Collapse
Affiliation(s)
- Madeline G Andrews
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Lakshmi Subramanian
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Pharmacology, Ideaya Biosciences, South San Francisco, CA, USA
| | - Jahan Salma
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Arnold R Kriegstein
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
17
|
Fischer J, Fernández Ortuño E, Marsoner F, Artioli A, Peters J, Namba T, Eugster Oegema C, Huttner WB, Ladewig J, Heide M. Human-specific ARHGAP11B ensures human-like basal progenitor levels in hominid cerebral organoids. EMBO Rep 2022; 23:e54728. [PMID: 36098218 PMCID: PMC9646322 DOI: 10.15252/embr.202254728] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/18/2022] [Accepted: 08/23/2022] [Indexed: 02/06/2023] Open
Abstract
The human-specific gene ARHGAP11B has been implicated in human neocortex expansion. However, the extent of ARHGAP11B's contribution to this expansion during hominid evolution is unknown. Here we address this issue by genetic manipulation of ARHGAP11B levels and function in chimpanzee and human cerebral organoids. ARHGAP11B expression in chimpanzee cerebral organoids doubles basal progenitor levels, the class of cortical progenitors with a key role in neocortex expansion. Conversely, interference with ARHGAP11B's function in human cerebral organoids decreases basal progenitors down to the chimpanzee level. Moreover, ARHGAP11A or ARHGAP11B rescue experiments in ARHGAP11A plus ARHGAP11B double-knockout human forebrain organoids indicate that lack of ARHGAP11B, but not of ARHGAP11A, decreases the abundance of basal radial glia-the basal progenitor type thought to be of particular relevance for neocortex expansion. Taken together, our findings demonstrate that ARHGAP11B is necessary and sufficient to ensure the elevated basal progenitor levels that characterize the fetal human neocortex, suggesting that this human-specific gene was a major contributor to neocortex expansion during human evolution.
Collapse
Affiliation(s)
- Jan Fischer
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Institute for Clinical GeneticsUniversity Hospital Carl Gustav CarusDresdenGermany
| | | | - Fabio Marsoner
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Annasara Artioli
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- Present address:
Neuroscience Center, HiLIFE ‐ Helsinki Institute of Life ScienceUniversity of HelsinkiHelsinkiFinland
| | | | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
| | - Julia Ladewig
- Central Institute of Mental HealthUniversity of Heidelberg/Medical Faculty MannheimMannheimGermany
- Hector Institute for Translational Brain Research (HITBR gGmbH)MannheimGermany
- German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Michael Heide
- Max Planck Institute of Molecular Cell Biology and GeneticsPfotenhauerstrasse 108DresdenGermany
- German Primate CenterLeibniz Institute for Primate ResearchGöttingenGermany
| |
Collapse
|
18
|
A kinase-independent function of cyclin-dependent kinase 6 promotes outer radial glia expansion and neocortical folding. Proc Natl Acad Sci U S A 2022; 119:e2206147119. [PMID: 36095192 PMCID: PMC9499540 DOI: 10.1073/pnas.2206147119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The neocortex, the center for higher brain function, first emerged in mammals and has become massively expanded and folded in humans, constituting almost half the volume of the human brain. Primary microcephaly, a developmental disorder in which the brain is smaller than normal at birth, results mainly from there being fewer neurons in the neocortex because of defects in neural progenitor cells (NPCs). Outer radial glia (oRGs), NPCs that are abundant in gyrencephalic species but rare in lissencephalic species, are thought to play key roles in the expansion and folding of the neocortex. However, how oRGs expand, whether they are necessary for neocortical folding, and whether defects in oRGs cause microcephaly remain important questions in the study of brain development, evolution, and disease. Here, we show that oRG expansion in mice, ferrets, and human cerebral organoids requires cyclin-dependent kinase 6 (CDK6), the mutation of which causes primary microcephaly via an unknown mechanism. In a mouse model in which increased Hedgehog signaling expands oRGs and intermediate progenitor cells and induces neocortical folding, CDK6 loss selectively decreased oRGs and abolished neocortical folding. Remarkably, this function of CDK6 in oRG expansion did not require its kinase activity, was not shared by the highly similar CDK4 and CDK2, and was disrupted by the mutation causing microcephaly. Therefore, our results indicate that CDK6 is conserved to promote oRG expansion, that oRGs are necessary for neocortical folding, and that defects in oRG expansion may cause primary microcephaly.
Collapse
|
19
|
Pinson A, Xing L, Namba T, Kalebic N, Peters J, Oegema CE, Traikov S, Reppe K, Riesenberg S, Maricic T, Derihaci R, Wimberger P, Pääbo S, Huttner WB. Human TKTL1 implies greater neurogenesis in frontal neocortex of modern humans than Neanderthals. Science 2022; 377:eabl6422. [PMID: 36074851 DOI: 10.1126/science.abl6422] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neanderthal brains were similar in size to those of modern humans. We sought to investigate potential differences in neurogenesis during neocortex development. Modern human transketolase-like 1 (TKTL1) differs from Neanderthal TKTL1 by a lysine-to-arginine amino acid substitution. Using overexpression in developing mouse and ferret neocortex, knockout in fetal human neocortical tissue, and genome-edited cerebral organoids, we found that the modern human variant, hTKTL1, but not the Neanderthal variant, increases the abundance of basal radial glia (bRG) but not that of intermediate progenitors (bIPs). bRG generate more neocortical neurons than bIPs. The hTKTL1 effect requires the pentose phosphate pathway and fatty acid synthesis. Inhibition of these metabolic pathways reduces bRG abundance in fetal human neocortical tissue. Our data suggest that neocortical neurogenesis in modern humans differs from that in Neanderthals.
Collapse
Affiliation(s)
- Anneline Pinson
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | | | - Sofia Traikov
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Katrin Reppe
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Stephan Riesenberg
- Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Tomislav Maricic
- Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Razvan Derihaci
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, 01307 Dresden, Germany
| | - Pauline Wimberger
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, 01307 Dresden, Germany
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
20
|
Brault J, Bardin S, Lampic M, Carpentieri JA, Coquand L, Penisson M, Lachuer H, Victoria GS, Baloul S, El Marjou F, Boncompain G, Miserey‐Lenkei S, Belvindrah R, Fraisier V, Francis F, Perez F, Goud B, Baffet AD. RAB6
and dynein drive
post‐Golgi
apical transport to prevent neuronal progenitor delamination. EMBO Rep 2022; 23:e54605. [PMID: 35979738 PMCID: PMC9535803 DOI: 10.15252/embr.202254605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 12/03/2022] Open
Abstract
Radial glial (RG) cells are the neural stem cells of the developing neocortex. Apical RG (aRG) cells can delaminate to generate basal RG (bRG) cells, a cell type associated with human brain expansion. Here, we report that aRG delamination is regulated by the post‐Golgi secretory pathway. Using in situ subcellular live imaging, we show that post‐Golgi transport of RAB6+ vesicles occurs toward the minus ends of microtubules and depends on dynein. We demonstrate that the apical determinant Crumbs3 (CRB3) is also transported by dynein. Double knockout of RAB6A/A' and RAB6B impairs apical localization of CRB3 and induces a retraction of aRG cell apical process, leading to delamination and ectopic division. These defects are phenocopied by knockout of the dynein activator LIS1. Overall, our results identify a RAB6‐dynein‐LIS1 complex for Golgi to apical surface transport in aRG cells, and highlights the role of this pathway in the maintenance of neuroepithelial integrity.
Collapse
Affiliation(s)
| | - Sabine Bardin
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Marusa Lampic
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | | | - Laure Coquand
- Institut Curie PSL Research University, CNRS UMR144 Paris France
- Sorbonne University Paris France
| | - Maxime Penisson
- Sorbonne University Paris France
- INSERM UMR‐S 1270 Paris France
- Institut du Fer à Moulin Paris France
| | - Hugo Lachuer
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | | | - Sarah Baloul
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Fatima El Marjou
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | | | | | - Richard Belvindrah
- Sorbonne University Paris France
- INSERM UMR‐S 1270 Paris France
- Institut du Fer à Moulin Paris France
| | - Vincent Fraisier
- UMR 144‐Cell and Tissue Imaging Facility (PICT‐IBiSA) CNRS‐Institut Curie Paris France
| | - Fiona Francis
- Sorbonne University Paris France
- INSERM UMR‐S 1270 Paris France
- Institut du Fer à Moulin Paris France
| | - Franck Perez
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Bruno Goud
- Institut Curie PSL Research University, CNRS UMR144 Paris France
| | - Alexandre D Baffet
- Institut Curie PSL Research University, CNRS UMR144 Paris France
- Institut National de la Santé et de la Recherche Médicale (INSERM) Paris France
| |
Collapse
|
21
|
Farcy S, Albert A, Gressens P, Baffet AD, El Ghouzzi V. Cortical Organoids to Model Microcephaly. Cells 2022; 11:2135. [PMID: 35883578 PMCID: PMC9320662 DOI: 10.3390/cells11142135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
How the brain develops and achieves its final size is a fascinating issue that questions cortical evolution across species and man's place in the animal kingdom. Although animal models have so far been highly valuable in understanding the key steps of cortical development, many human specificities call for appropriate models. In particular, microcephaly, a neurodevelopmental disorder that is characterized by a smaller head circumference has been challenging to model in mice, which often do not fully recapitulate the human phenotype. The relatively recent development of brain organoid technology from induced pluripotent stem cells (iPSCs) now makes it possible to model human microcephaly, both due to genetic and environmental origins, and to generate developing cortical tissue from the patients themselves. These 3D tissues rely on iPSCs differentiation into cortical progenitors that self-organize into neuroepithelial rosettes mimicking the earliest stages of human neurogenesis in vitro. Over the last ten years, numerous protocols have been developed to control the identity of the induced brain areas, the reproducibility of the experiments and the longevity of the cultures, allowing analysis of the later stages. In this review, we describe the different approaches that instruct human iPSCs to form cortical organoids, summarize the different microcephalic conditions that have so far been modeled by organoids, and discuss the relevance of this model to decipher the cellular and molecular mechanisms of primary and secondary microcephalies.
Collapse
Affiliation(s)
- Sarah Farcy
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Alexandra Albert
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Pierre Gressens
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Alexandre D. Baffet
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Vincent El Ghouzzi
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| |
Collapse
|
22
|
Espinós A, Fernández‐Ortuño E, Negri E, Borrell V. Evolution of genetic mechanisms regulating cortical neurogenesis. Dev Neurobiol 2022; 82:428-453. [PMID: 35670518 PMCID: PMC9543202 DOI: 10.1002/dneu.22891] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/26/2022] [Accepted: 05/24/2022] [Indexed: 11/20/2022]
Abstract
The size of the cerebral cortex increases dramatically across amniotes, from reptiles to great apes. This is primarily due to different numbers of neurons and glial cells produced during embryonic development. The evolutionary expansion of cortical neurogenesis was linked to changes in neural stem and progenitor cells, which acquired increased capacity of self‐amplification and neuron production. Evolution works via changes in the genome, and recent studies have identified a small number of new genes that emerged in the recent human and primate lineages, promoting cortical progenitor proliferation and increased neurogenesis. However, most of the mammalian genome corresponds to noncoding DNA that contains gene‐regulatory elements, and recent evidence precisely points at changes in expression levels of conserved genes as key in the evolution of cortical neurogenesis. Here, we provide an overview of basic cellular mechanisms involved in cortical neurogenesis across amniotes, and discuss recent progress on genetic mechanisms that may have changed during evolution, including gene expression regulation, leading to the expansion of the cerebral cortex.
Collapse
Affiliation(s)
- Alexandre Espinós
- Instituto de Neurociencias CSIC ‐ UMH, 03550 Sant Joan d'Alacant Spain
| | | | - Enrico Negri
- Instituto de Neurociencias CSIC ‐ UMH, 03550 Sant Joan d'Alacant Spain
| | - Víctor Borrell
- Instituto de Neurociencias CSIC ‐ UMH, 03550 Sant Joan d'Alacant Spain
| |
Collapse
|
23
|
Vaid S, Huttner WB. Progenitor-Based Cell Biological Aspects of Neocortex Development and Evolution. Front Cell Dev Biol 2022; 10:892922. [PMID: 35602606 PMCID: PMC9119302 DOI: 10.3389/fcell.2022.892922] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
During development, the decision of stem and progenitor cells to switch from proliferation to differentiation is of critical importance for the overall size of an organ. Too early a switch will deplete the stem/progenitor cell pool, and too late a switch will not generate the required differentiated cell types. With a focus on the developing neocortex, a six-layered structure constituting the major part of the cerebral cortex in mammals, we discuss here the cell biological features that are crucial to ensure the appropriate proliferation vs. differentiation decision in the neural progenitor cells. In the last two decades, the neural progenitor cells giving rise to the diverse types of neurons that function in the neocortex have been intensely investigated for their role in cortical expansion and gyrification. In this review, we will first describe these different progenitor types and their diversity. We will then review the various cell biological features associated with the cell fate decisions of these progenitor cells, with emphasis on the role of the radial processes emanating from these progenitor cells. We will also discuss the species-specific differences in these cell biological features that have allowed for the evolutionary expansion of the neocortex in humans. Finally, we will discuss the emerging role of cell cycle parameters in neocortical expansion.
Collapse
Affiliation(s)
- Samir Vaid
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- *Correspondence: Samir Vaid, ; Wieland B. Huttner,
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- *Correspondence: Samir Vaid, ; Wieland B. Huttner,
| |
Collapse
|
24
|
Shinmyo Y, Hamabe-Horiike T, Saito K, Kawasaki H. Investigation of the Mechanisms Underlying the Development and Evolution of the Cerebral Cortex Using Gyrencephalic Ferrets. Front Cell Dev Biol 2022; 10:847159. [PMID: 35386196 PMCID: PMC8977464 DOI: 10.3389/fcell.2022.847159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex has changed significantly during evolution. As a result of the increase in the number of neurons and glial cells in the cerebral cortex, its size has markedly expanded. Moreover, folds, called gyri and sulci, appeared on its surface, and its neuronal circuits have become much more complicated. Although these changes during evolution are considered to have been crucial for the acquisition of higher brain functions, the mechanisms underlying the development and evolution of the cerebral cortex of mammals are still unclear. This is, at least partially, because it is difficult to investigate these mechanisms using mice only. Therefore, genetic manipulation techniques for the cerebral cortex of gyrencephalic carnivore ferrets were developed recently. Furthermore, gene knockout was achieved in the ferret cerebral cortex using the CRISPR/Cas9 system. These techniques enabled molecular investigations using the ferret cerebral cortex. In this review, we will summarize recent findings regarding the mechanisms underlying the development and evolution of the mammalian cerebral cortex, mainly focusing on research using ferrets.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
25
|
Pathophysiological Heterogeneity of the BBSOA Neurodevelopmental Syndrome. Cells 2022; 11:cells11081260. [PMID: 35455940 PMCID: PMC9024734 DOI: 10.3390/cells11081260] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
The formation and maturation of the human brain is regulated by highly coordinated developmental events, such as neural cell proliferation, migration and differentiation. Any impairment of these interconnected multi-factorial processes can affect brain structure and function and lead to distinctive neurodevelopmental disorders. Here, we review the pathophysiology of the Bosch–Boonstra–Schaaf Optic Atrophy Syndrome (BBSOAS; OMIM 615722; ORPHA 401777), a recently described monogenic neurodevelopmental syndrome caused by the haploinsufficiency of NR2F1 gene, a key transcriptional regulator of brain development. Although intellectual disability, developmental delay and visual impairment are arguably the most common symptoms affecting BBSOAS patients, multiple additional features are often reported, including epilepsy, autistic traits and hypotonia. The presence of specific symptoms and their variable level of severity might depend on still poorly characterized genotype–phenotype correlations. We begin with an overview of the several mutations of NR2F1 identified to date, then further focuses on the main pathological features of BBSOAS patients, providing evidence—whenever possible—for the existing genotype–phenotype correlations. On the clinical side, we lay out an up-to-date list of clinical examinations and therapeutic interventions recommended for children with BBSOAS. On the experimental side, we describe state-of-the-art in vivo and in vitro studies aiming at deciphering the role of mouse Nr2f1, in physiological conditions and in pathological contexts, underlying the BBSOAS features. Furthermore, by modeling distinct NR2F1 genetic alterations in terms of dimer formation and nuclear receptor binding efficiencies, we attempt to estimate the total amounts of functional NR2F1 acting in developing brain cells in normal and pathological conditions. Finally, using the NR2F1 gene and BBSOAS as a paradigm of monogenic rare neurodevelopmental disorder, we aim to set the path for future explorations of causative links between impaired brain development and the appearance of symptoms in human neurological syndromes.
Collapse
|
26
|
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
|
27
|
Salamon I, Rasin MR. Evolution of the Neocortex Through RNA-Binding Proteins and Post-transcriptional Regulation. Front Neurosci 2022; 15:803107. [PMID: 35082597 PMCID: PMC8784817 DOI: 10.3389/fnins.2021.803107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
The human neocortex is undoubtedly considered a supreme accomplishment in mammalian evolution. It features a prenatally established six-layered structure which remains plastic to the myriad of changes throughout an organism’s lifetime. A fundamental feature of neocortical evolution and development is the abundance and diversity of the progenitor cell population and their neuronal and glial progeny. These evolutionary upgrades are partially enabled due to the progenitors’ higher proliferative capacity, compartmentalization of proliferative regions, and specification of neuronal temporal identities. The driving force of these processes may be explained by temporal molecular patterning, by which progenitors have intrinsic capacity to change their competence as neocortical neurogenesis proceeds. Thus, neurogenesis can be conceptualized along two timescales of progenitors’ capacity to (1) self-renew or differentiate into basal progenitors (BPs) or neurons or (2) specify their fate into distinct neuronal and glial subtypes which participate in the formation of six-layers. Neocortical development then proceeds through sequential phases of proliferation, differentiation, neuronal migration, and maturation. Temporal molecular patterning, therefore, relies on the precise regulation of spatiotemporal gene expression. An extensive transcriptional regulatory network is accompanied by post-transcriptional regulation that is frequently mediated by the regulatory interplay between RNA-binding proteins (RBPs). RBPs exhibit important roles in every step of mRNA life cycle in any system, from splicing, polyadenylation, editing, transport, stability, localization, to translation (protein synthesis). Here, we underscore the importance of RBP functions at multiple time-restricted steps of early neurogenesis, starting from the cell fate transition of transcriptionally primed cortical progenitors. A particular emphasis will be placed on RBPs with mostly conserved but also divergent evolutionary functions in neural progenitors across different species. RBPs, when considered in the context of the fascinating process of neocortical development, deserve to be main protagonists in the story of the evolution and development of the neocortex.
Collapse
|
28
|
Kyrousi C, O’Neill AC, Brazovskaja A, He Z, Kielkowski P, Coquand L, Di Giaimo R, D’ Andrea P, Belka A, Forero Echeverry A, Mei D, Lenge M, Cruceanu C, Buchsbaum IY, Khattak S, Fabien G, Binder E, Elmslie F, Guerrini R, Baffet AD, Sieber SA, Treutlein B, Robertson SP, Cappello S. Extracellular LGALS3BP regulates neural progenitor position and relates to human cortical complexity. Nat Commun 2021; 12:6298. [PMID: 34728600 PMCID: PMC8564519 DOI: 10.1038/s41467-021-26447-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 09/26/2021] [Indexed: 12/15/2022] Open
Abstract
Basal progenitors (BPs), including intermediate progenitors and basal radial glia, are generated from apical radial glia and are enriched in gyrencephalic species like humans, contributing to neuronal expansion. Shortly after generation, BPs delaminate towards the subventricular zone, where they further proliferate before differentiation. Gene expression alterations involved in BP delamination and function in humans are poorly understood. Here, we study the role of LGALS3BP, so far known as a cancer biomarker, which is a secreted protein enriched in human neural progenitors (NPCs). We show that individuals with LGALS3BP de novo variants exhibit altered local gyrification, sulcal depth, surface area and thickness in their cortex. Additionally, using cerebral organoids, human fetal tissues and mice, we show that LGALS3BP regulates the position of NPCs. Single-cell RNA-sequencing and proteomics reveal that LGALS3BP-mediated mechanisms involve the extracellular matrix in NPCs' anchoring and migration within the human brain. We propose that its temporal expression influences NPCs' delamination, corticogenesis and gyrification extrinsically.
Collapse
Affiliation(s)
- Christina Kyrousi
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany ,grid.5216.00000 0001 2155 0800Present Address: First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Greece and University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
| | - Adam C. O’Neill
- grid.29980.3a0000 0004 1936 7830Department of Women’s and Children’s Health, University of Otago, 9054 Dunedin, New Zealand
| | - Agnieska Brazovskaja
- grid.419518.00000 0001 2159 1813Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Zhisong He
- grid.419518.00000 0001 2159 1813Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany ,grid.5801.c0000 0001 2156 2780ETH Zurich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| | - Pavel Kielkowski
- grid.6936.a0000000123222966Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany ,grid.5252.00000 0004 1936 973XPresent Address: Department Chemie Ludwig-Maximilians-Universität München Butenandtstr. 5-13, 81377 München, Germany
| | - Laure Coquand
- grid.4444.00000 0001 2112 9282Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d’Ulm, F-75005 Paris, France
| | - Rossella Di Giaimo
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany ,grid.4691.a0000 0001 0790 385XDepartment of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Pierpaolo D’ Andrea
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alexander Belka
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | - Davide Mei
- grid.413181.e0000 0004 1757 8562Neuroscience Department, Children’s Hospital A. Meyer-University of Florence, 50139 Florence, Italy
| | - Matteo Lenge
- grid.413181.e0000 0004 1757 8562Neuroscience Department, Children’s Hospital A. Meyer-University of Florence, 50139 Florence, Italy
| | - Cristiana Cruceanu
- grid.419548.50000 0000 9497 5095Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Isabel Y. Buchsbaum
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany ,grid.5252.00000 0004 1936 973XGraduate School of Systemic Neurosciences, Ludwig-Maximilians-University, 82152 Munich Planegg, Germany
| | - Shahryar Khattak
- grid.4488.00000 0001 2111 7257DFG-Research Center and Cluster of Excellence for Regenerative Therapies (CRTD), School of Medicine, Technical University Dresden, 01307 Dresden, Germany ,grid.4912.e0000 0004 0488 7120Present Address: Royal College of Surgeons Ireland (RCSI) in Bahrain, Adliya, Kingdom of Bahrain
| | - Guimiot Fabien
- grid.50550.350000 0001 2175 4109Unité de Foetopathologie, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, F-75019 Paris, France
| | - Elisabeth Binder
- grid.419548.50000 0000 9497 5095Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Frances Elmslie
- grid.4464.20000 0001 2161 2573South West Thames Regional Genetics Service, St George’s, University of London, London, SW17 0RE UK
| | - Renzo Guerrini
- grid.413181.e0000 0004 1757 8562Neuroscience Department, Children’s Hospital A. Meyer-University of Florence, 50139 Florence, Italy
| | - Alexandre D. Baffet
- grid.4444.00000 0001 2112 9282Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d’Ulm, F-75005 Paris, France
| | - Stephan A. Sieber
- grid.6936.a0000000123222966Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany
| | - Barbara Treutlein
- grid.419518.00000 0001 2159 1813Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany ,grid.5801.c0000 0001 2156 2780ETH Zurich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| | - Stephen P. Robertson
- grid.29980.3a0000 0004 1936 7830Department of Women’s and Children’s Health, University of Otago, 9054 Dunedin, New Zealand
| | - Silvia Cappello
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
29
|
Kerimoglu C, Pham L, Tonchev AB, Sakib MS, Xie Y, Sokpor G, Ulmke PA, Kaurani L, Abbas E, Nguyen H, Rosenbusch J, Michurina A, Capece V, Angelova M, Maricic N, Brand-Saberi B, Esgleas M, Albert M, Minkov R, Kovachev E, Teichmann U, Seong RH, Huttner WB, Nguyen HP, Stoykova A, Staiger JF, Fischer A, Tuoc T. H3 acetylation selectively promotes basal progenitor proliferation and neocortex expansion. SCIENCE ADVANCES 2021; 7:eabc6792. [PMID: 34524839 PMCID: PMC8443185 DOI: 10.1126/sciadv.abc6792] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/23/2021] [Indexed: 06/13/2023]
Abstract
Increase in the size of human neocortex―acquired in evolution―accounts for the unique cognitive capacity of humans. This expansion reflects the evolutionarily enhanced proliferative ability of basal progenitors (BPs), including the basal radial glia and basal intermediate progenitors (bIPs) in mammalian cortex, which may have been acquired through epigenetic alterations in BPs. However, how the epigenome in BPs differs across species is not known. Here, we report that histone H3 acetylation is a key epigenetic regulation in bIP amplification and cortical expansion. Through epigenetic profiling of sorted bIPs, we show that histone H3 lysine 9 acetylation (H3K9ac) is low in murine bIPs and high in human bIPs. Elevated H3K9ac preferentially increases bIP proliferation, increasing the size and folding of the normally smooth mouse neocortex. H3K9ac drives bIP amplification by increasing expression of the evolutionarily regulated gene, Trnp1, in developing cortex. Our findings demonstrate a previously unknown mechanism that controls cortical architecture.
Collapse
Affiliation(s)
- Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Linh Pham
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Anton B. Tonchev
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
- Departments of Anatomy and Cell Biology and Stem Cell Biology, Research Institute, Medical University of Varna, Varna 9002, Bulgaria
| | - M. Sadman Sakib
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Yuanbin Xie
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Gannan Medical University, Ganzhou 341000, The People’s Republic of China
| | - Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Pauline Antonie Ulmke
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Lalit Kaurani
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Eman Abbas
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Huong Nguyen
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Faculty of Biotechnology, Thai Nguyen University of Sciences, Thai Nguyen, Vietnam
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | | | - Vincenzo Capece
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | - Meglena Angelova
- Departments of Anatomy and Cell Biology and Stem Cell Biology, Research Institute, Medical University of Varna, Varna 9002, Bulgaria
| | - Nenad Maricic
- Institute of Anatomy and Molecular Embryology, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Institute of Anatomy and Molecular Embryology, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Miriam Esgleas
- Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Mareike Albert
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Radoslav Minkov
- Specialized Hospital for Obstetrics and Gynecology “Prof. Dimitar Stamatov” –Varna, Medical University of Varna, Varna 9002, Bulgaria
| | - Emil Kovachev
- Specialized Hospital for Obstetrics and Gynecology “Prof. Dimitar Stamatov” –Varna, Medical University of Varna, Varna 9002, Bulgaria
| | - Ulrike Teichmann
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
| | - Rho H. Seong
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Anastassia Stoykova
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| | - Jochen F. Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| |
Collapse
|
30
|
Yang L, Li Z, Liu G, Li X, Yang Z. Developmental Origins of Human Cortical Oligodendrocytes and Astrocytes. Neurosci Bull 2021; 38:47-68. [PMID: 34374948 PMCID: PMC8783027 DOI: 10.1007/s12264-021-00759-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/27/2021] [Indexed: 12/30/2022] Open
Abstract
Human cortical radial glial cells are primary neural stem cells that give rise to cortical glutaminergic projection pyramidal neurons, glial cells (oligodendrocytes and astrocytes) and olfactory bulb GABAergic interneurons. One of prominent features of the human cortex is enriched with glial cells, but there are major gaps in understanding how these glial cells are generated. Herein, by integrating analysis of published human cortical single-cell RNA-Seq datasets with our immunohistochemistical analyses, we show that around gestational week 18, EGFR-expressing human cortical truncated radial glial cells (tRGs) give rise to basal multipotent intermediate progenitors (bMIPCs) that express EGFR, ASCL1, OLIG2 and OLIG1. These bMIPCs undergo several rounds of mitosis and generate cortical oligodendrocytes, astrocytes and olfactory bulb interneurons. We also characterized molecular features of the cortical tRG. Integration of our findings suggests a general picture of the lineage progression of cortical radial glial cells, a fundamental process of the developing human cerebral cortex.
Collapse
Affiliation(s)
- Lin Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institute for Translational Brain Research, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhenmeiyu Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institute for Translational Brain Research, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institute for Translational Brain Research, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaosu Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institute for Translational Brain Research, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Institute for Translational Brain Research, and Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
31
|
Tatomir A, Beltrand A, Nguyen V, Courneya JP, Boodhoo D, Cudrici C, Muresanu DF, Rus V, Badea TC, Rus H. RGC-32 Acts as a Hub to Regulate the Transcriptomic Changes Associated With Astrocyte Development and Reactive Astrocytosis. Front Immunol 2021; 12:705308. [PMID: 34394104 PMCID: PMC8358671 DOI: 10.3389/fimmu.2021.705308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/16/2021] [Indexed: 01/14/2023] Open
Abstract
Response Gene to Complement 32 (RGC-32) is an important mediator of the TGF-β signaling pathway, and an increasing amount of evidence implicates this protein in regulating astrocyte biology. We showed recently that spinal cord astrocytes in mice lacking RGC-32 display an immature phenotype reminiscent of progenitors and radial glia, with an overall elongated morphology, increased proliferative capacity, and increased expression of progenitor markers when compared to their wild-type (WT) counterparts that make them incapable of undergoing reactive changes during the acute phase of experimental autoimmune encephalomyelitis (EAE). Here, in order to decipher the molecular networks underlying RGC-32's ability to regulate astrocytic maturation and reactivity, we performed next-generation sequencing of RNA from WT and RGC-32 knockout (KO) neonatal mouse brain astrocytes, either unstimulated or stimulated with the pleiotropic cytokine TGF-β. Pathway enrichment analysis showed that RGC-32 is critical for the TGF-β-induced up-regulation of transcripts encoding proteins involved in brain development and tissue remodeling, such as axonal guidance molecules, transcription factors, extracellular matrix (ECM)-related proteins, and proteoglycans. Our next-generation sequencing of RNA analysis also demonstrated that a lack of RGC-32 results in a significant induction of WD repeat and FYVE domain-containing protein 1 (Wdfy1) and stanniocalcin-1 (Stc1). Immunohistochemical analysis of spinal cords isolated from normal adult mice and mice with EAE at the peak of disease showed that RGC-32 is necessary for the in vivo expression of ephrin receptor type A7 in reactive astrocytes, and that the lack of RGC-32 results in a higher number of homeodomain-only protein homeobox (HOPX)+ and CD133+ radial glia cells. Collectively, these findings suggest that RGC-32 plays a major role in modulating the transcriptomic changes in astrocytes that ultimately lead to molecular programs involved in astrocytic differentiation and reactive changes during neuroinflammation.
Collapse
Affiliation(s)
- Alexandru Tatomir
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Austin Beltrand
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Vinh Nguyen
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Jean-Paul Courneya
- Health Sciences and Human Services Library, University of Maryland, Baltimore, MD, United States
| | - Dallas Boodhoo
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Cornelia Cudrici
- Translational Vascular Medicine Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Dafin F. Muresanu
- Department of Neurosciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Violeta Rus
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Tudor C. Badea
- Retinal Circuit Development and Genetics Unit, N-NRL, National Eye Institute, Bethesda, MD, United States
- Research and Development Institute, Faculty of Medicine, Transylvania University of Brasov, Brasov, Romania
| | - Horea Rus
- Department of Neurology, University of Maryland, School of Medicine, Baltimore, MD, United States
- Research Service, Veterans Administration Maryland Health Care System, Baltimore, MD, United States
| |
Collapse
|
32
|
HOPX Exhibits Oncogenic Activity during Squamous Skin Carcinogenesis. J Invest Dermatol 2021; 141:2354-2368. [PMID: 33845078 DOI: 10.1016/j.jid.2020.04.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 11/23/2022]
Abstract
Cutaneous squamous cell carcinomas (SCCs) are frequent heterogeneous tumors arising from sun-exposed regions of the skin and characterized by complex pathogenesis. HOPX is a member of the homeodomain-containing superfamily of proteins holding an atypical homeodomain unable to bind to DNA. First discovered in the heart as a regulator of cardiac development, in the skin, HOPX modulates the terminal differentiation of keratinocytes. There is a particular interest in studying HOPX in squamous skin carcinogenesis because it has the atypical structure and the functional duality as an oncogene and a tumor suppressor gene, reported in different malignancies. In this study, we analyzed the effects of HOPX knockdown and overexpression on SCC tumorigenicity in vitro and in vivo. Our data show that HOPX knockdown in SCC cells inhibits their proliferative and invasive activity through the acceleration of apoptosis. We established that methylation of two alternative HOPX promoters leads to differential expression of HOPX transcripts in normal keratinocytes and SCC cells. Importantly, we report that HOPX acts as an oncogene in the pathogenesis of SCC probably through the activation of the second alternative promoter and the modulation of apoptosis.
Collapse
|
33
|
D'Souza L, Channakkar AS, Muralidharan B. Chromatin remodelling complexes in cerebral cortex development and neurodevelopmental disorders. Neurochem Int 2021; 147:105055. [PMID: 33964373 PMCID: PMC7611358 DOI: 10.1016/j.neuint.2021.105055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/11/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022]
Abstract
The diverse number of neurons in the cerebral cortex are generated during development by neural stem cells lining the ventricle, and they continue maturing postnatally. Dynamic chromatin regulation in these neural stem cells is a fundamental determinant of the emerging property of the functional neural network, and the chromatin remodellers are critical determinants of this process. Chromatin remodellers participate in several steps of this process from proliferation, differentiation, migration leading to complex network formation which forms the basis of higher-order functions of cognition and behaviour. Here we review the role of these ATP-dependent chromatin remodellers in cortical development in health and disease and highlight several key mouse mutants of the subunits of the complexes which have revealed how the remodelling mechanisms control the cortical stem cell chromatin landscape for expression of stage-specific transcripts. Consistent with their role in cortical development, several putative risk variants in the subunits of the remodelling complexes have been identified as the underlying causes of several neurodevelopmental disorders. A basic understanding of the detailed molecular mechanism of their action is key to understating how mutations in the same networks lead to disease pathologies and perhaps pave the way for therapeutic development for these complex multifactorial disorders.
Collapse
Affiliation(s)
- Leora D'Souza
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Asha S Channakkar
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India
| | - Bhavana Muralidharan
- Brain Development and Disease Mechanisms, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore Life Science Cluster, Bangalore, India.
| |
Collapse
|
34
|
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: 28] [Impact Index Per Article: 7.0] [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
|
35
|
Yoshinaga S, Shin M, Kitazawa A, Ishii K, Tanuma M, Kasai A, Hashimoto H, Kubo KI, Nakajima K. Comprehensive characterization of migration profiles of murine cerebral cortical neurons during development using FlashTag labeling. iScience 2021; 24:102277. [PMID: 33851097 PMCID: PMC8022222 DOI: 10.1016/j.isci.2021.102277] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/30/2020] [Accepted: 03/01/2021] [Indexed: 11/26/2022] Open
Abstract
In the mammalian cerebral neocortex, different regions have different cytoarchitecture, neuronal birthdates, and functions. In most regions, neuronal migratory profiles are speculated similar based on observations using thymidine analogs. Few reports have investigated regional migratory differences from mitosis at the ventricular surface. In this study, we applied FlashTag technology, in which dyes are injected intraventricularly, to describe migratory profiles. We revealed a mediolateral regional difference in the migratory profiles of neurons that is dependent on developmental stage; for example, neurons labeled at embryonic day 12.5–15.5 reached their destination earlier dorsomedially than dorsolaterally, even where there were underlying ventricular surfaces, reflecting sojourning below the subplate. This difference was hardly recapitulated by thymidine analogs, which visualize neurogenic gradients, suggesting a biological significance different from the neurogenic gradient. These observations advance our understanding of cortical development and the power of FlashTag in studying migration and are thus resources for future neurodevelopmental studies. FlashTag visualized mediolateral regional differences of cortical migratory profiles Mediolateral differences were observed when neurons were labeled at E12.5–15.5 Late-born neurons transiently sojourned below the dorsolateral subplate (SP) cells The difference was unclear in reeler cortex, where SP cells position superficially
Collapse
Affiliation(s)
- Satoshi Yoshinaga
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Minkyung Shin
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Ayako Kitazawa
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Masato Tanuma
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsushi Kasai
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan
| | - Hitoshi Hashimoto
- Laboratory of Molecular Neuropharmacology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka 565-0871, Japan.,Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University, and University of Fukui, Suita, Osaka 565-0871, Japan.,Division of Bioscience, Institute for Datability Science, Osaka University, Suita, Osaka 565-0871, Japan.,Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan.,Department of Molecular Pharmaceutical Sciences, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan.,Department of Anatomy, The Jikei University School of Medicine, Minato, Tokyo 105-8461, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
36
|
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
|
37
|
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: 53] [Impact Index Per Article: 10.6] [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
|
38
|
Farooq M, Lindbæk L, Krogh N, Doganli C, Keller C, Mönnich M, Gonçalves AB, Sakthivel S, Mang Y, Fatima A, Andersen VS, Hussain MS, Eiberg H, Hansen L, Kjaer KW, Gopalakrishnan J, Pedersen LB, Møllgård K, Nielsen H, Baig SM, Tommerup N, Christensen ST, Larsen LA. RRP7A links primary microcephaly to dysfunction of ribosome biogenesis, resorption of primary cilia, and neurogenesis. Nat Commun 2020; 11:5816. [PMID: 33199730 PMCID: PMC7670429 DOI: 10.1038/s41467-020-19658-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/26/2020] [Indexed: 12/25/2022] Open
Abstract
Primary microcephaly (MCPH) is characterized by reduced brain size and intellectual disability. The exact pathophysiological mechanism underlying MCPH remains to be elucidated, but dysfunction of neuronal progenitors in the developing neocortex plays a major role. We identified a homozygous missense mutation (p.W155C) in Ribosomal RNA Processing 7 Homolog A, RRP7A, segregating with MCPH in a consanguineous family with 10 affected individuals. RRP7A is highly expressed in neural stem cells in developing human forebrain, and targeted mutation of Rrp7a leads to defects in neurogenesis and proliferation in a mouse stem cell model. RRP7A localizes to centrosomes, cilia and nucleoli, and patient-derived fibroblasts display defects in ribosomal RNA processing, primary cilia resorption, and cell cycle progression. Analysis of zebrafish embryos supported that the patient mutation in RRP7A causes reduced brain size, impaired neurogenesis and cell proliferation, and defective ribosomal RNA processing. These findings provide novel insight into human brain development and MCPH. The RRP7A a gene is involved in ribosome biogenesis. Here the authors report a homozygous missense mutation segregating with primary microcephaly, and show that this occurs via functional defects in both nucleoli and primary cilia disrupting cell proliferation and neurogenesis.
Collapse
Affiliation(s)
- Muhammad Farooq
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark.,Department of Biochemistry and Biotechnology, The Islamia University of Bahawalpur, Baghdad ul Jadeed Campus, 63100, Bahawalpur, Punjab, Pakistan
| | - Louise Lindbæk
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Nicolai Krogh
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Canan Doganli
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Cecilie Keller
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Maren Mönnich
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - André Brás Gonçalves
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Srinivasan Sakthivel
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Yuan Mang
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Ambrin Fatima
- Human Molecular Genetics Laboratory; Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering PIEAS, Jhang Road, 38000, Faisalabad, Punjab, Pakistan
| | - Vivi Søgaard Andersen
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Muhammad S Hussain
- Institute of Biochemistry I, University of Cologne, Joseph-Stelzmann-Strasse 52, D50931, Cologne, Germany.,Cologne Center for Genomics and Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch-Strasse 21, D50931, Cologne, Germany
| | - Hans Eiberg
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Lars Hansen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Klaus Wilbrandt Kjaer
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Jay Gopalakrishnan
- Institute of Human Genetics, Universitätsstrasse 1, Heinrich-Heine-University, D-40225, Düsseldorf, Germany
| | - Lotte Bang Pedersen
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark
| | - Kjeld Møllgård
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Henrik Nielsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Shahid M Baig
- Human Molecular Genetics Laboratory; Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering PIEAS, Jhang Road, 38000, Faisalabad, Punjab, Pakistan
| | - Niels Tommerup
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark
| | - Søren Tvorup Christensen
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100, Copenhagen, Denmark.
| | - Lars Allan Larsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
39
|
Li Z, Tyler WA, Zeldich E, Santpere Baró G, Okamoto M, Gao T, Li M, Sestan N, Haydar TF. Transcriptional priming as a conserved mechanism of lineage diversification in the developing mouse and human neocortex. SCIENCE ADVANCES 2020; 6:6/45/eabd2068. [PMID: 33158872 PMCID: PMC7673705 DOI: 10.1126/sciadv.abd2068] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/23/2020] [Indexed: 05/23/2023]
Abstract
How the rich variety of neurons in the nervous system arises from neural stem cells is not well understood. Using single-cell RNA-sequencing and in vivo confirmation, we uncover previously unrecognized neural stem and progenitor cell diversity within the fetal mouse and human neocortex, including multiple types of radial glia and intermediate progenitors. We also observed that transcriptional priming underlies the diversification of a subset of ventricular radial glial cells in both species; genetic fate mapping confirms that the primed radial glial cells generate specific types of basal progenitors and neurons. The different precursor lineages therefore diversify streams of cell production in the developing murine and human neocortex. These data show that transcriptional priming is likely a conserved mechanism of mammalian neural precursor lineage specialization.
Collapse
Affiliation(s)
- Zhen Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - William A Tyler
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Ella Zeldich
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Gabriel Santpere Baró
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Neurogenomics group, Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), DCEXS, Universitat Pompeu Fabra, Barcelona, Spain
| | - Mayumi Okamoto
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Aichi 466-8550, Japan
| | - Tianliuyun Gao
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Mingfeng Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT, USA.
- Departments of Genetics, of Psychiatry and of Comparative Medicine, Program in Cellular Neuroscience, Neurodegeneration and Repair, Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Tarik F Haydar
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
40
|
Xing L, Kalebic N, Namba T, Vaid S, Wimberger P, Huttner WB. Serotonin Receptor 2A Activation Promotes Evolutionarily Relevant Basal Progenitor Proliferation in the Developing Neocortex. Neuron 2020; 108:1113-1129.e6. [PMID: 33080227 DOI: 10.1016/j.neuron.2020.09.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/17/2020] [Accepted: 09/24/2020] [Indexed: 12/26/2022]
Abstract
Evolutionary expansion of the mammalian neocortex (Ncx) has been linked to increased abundance and proliferative capacity of basal progenitors (BPs) in the subventricular zone during development. BP proliferation is governed by both intrinsic and extrinsic signals, several of which have been identified. However, a role of neurotransmitters, a canonical class of extrinsic signaling molecules, in BP proliferation remains to be established. Here, we show that serotonin (5-HT), via its receptor HTR2A, promotes BP proliferation in an evolutionarily relevant manner. HTR2A is not expressed in embryonic mouse Ncx; accordingly, 5-HT does not increase mouse BP proliferation. However, ectopic HTR2A expression can increase mouse BP proliferation. Conversely, CRISPR/Cas9-mediated knockout of endogenous HTR2A in embryonic ferret Ncx reduces BP proliferation. Pharmacological activation of endogenous HTR2A in fetal human Ncx ex vivo increases BP proliferation via HER2/ERK signaling. Hence, 5-HT emerges as an important extrinsic pro-proliferative signal for BPs, which may have contributed to evolutionary Ncx expansion.
Collapse
Affiliation(s)
- Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany; Human Technopole, Via Cristina Belgioioso 171, Milan, Italy
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Pauline Wimberger
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|
41
|
Stepien BK, Naumann R, Holtz A, Helppi J, Huttner WB, Vaid S. Lengthening Neurogenic Period during Neocortical Development Causes a Hallmark of Neocortex Expansion. Curr Biol 2020; 30:4227-4237.e5. [PMID: 32888487 DOI: 10.1016/j.cub.2020.08.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 07/09/2020] [Accepted: 08/12/2020] [Indexed: 02/07/2023]
Abstract
A hallmark of the evolutionary expansion of the neocortex is a specific increase in the number of neurons generated for the upper neocortical layers during development. The cause underlying this increase is unknown. Here, we show that lengthening the neurogenic period during neocortical development is sufficient to specifically increase upper-layer neuron generation. Thus, embryos of mouse strains with longer gestation exhibited a longer neurogenic period and generated more upper-layer, but not more deep-layer, neurons than embryos with shorter gestation. Accordingly, long-gestation embryos showed a greater abundance of neurogenic progenitors in the subventricular zone than short-gestation embryos at late stages of cortical neurogenesis. Analysis of a mouse-rat chimeric embryo, developing inside a rat mother, pointed to factors in the rat environment that influenced the upper-layer neuron generation by the mouse progenitors. Exploring a potential maternal source of such factors, short-gestation strain mouse embryos transferred to long-gestation strain mothers exhibited an increase in the length of the neurogenic period and upper-layer neuron generation. The opposite was the case for long-gestation strain mouse embryos transferred to short-gestation strain mothers, indicating a dominant maternal influence on the length of the neurogenic period and hence upper-layer neuron generation. In summary, our study uncovers a hitherto unknown link between embryonic cortical neurogenesis and the maternal gestational environment and provides experimental evidence that lengthening the neurogenic period during neocortical development underlies a key aspect of neocortical expansion.
Collapse
Affiliation(s)
- Barbara K Stepien
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Anja Holtz
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Jussi Helppi
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany.
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany.
| |
Collapse
|
42
|
Goodman T, Nayar SG, Clare S, Mikolajczak M, Rice R, Mansour S, Bellusci S, Hajihosseini MK. Fibroblast growth factor 10 is a negative regulator of postnatal neurogenesis in the mouse hypothalamus. Development 2020; 147:147/13/dev180950. [PMID: 32661019 PMCID: PMC7375484 DOI: 10.1242/dev.180950] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/03/2020] [Indexed: 12/13/2022]
Abstract
New neurons are generated in the postnatal rodent hypothalamus, with a subset of tanycytes in the third ventricular (3V) wall serving as neural stem/progenitor cells. However, the precise stem cell niche organization, the intermediate steps and the endogenous regulators of postnatal hypothalamic neurogenesis remain elusive. Quantitative lineage-tracing in vivo revealed that conditional deletion of fibroblast growth factor 10 (Fgf10) from Fgf10-expressing β-tanycytes at postnatal days (P)4-5 results in the generation of significantly more parenchymal cells by P28, composed mostly of ventromedial and dorsomedial neurons and some glial cells, which persist into adulthood. A closer scrutiny in vivo and ex vivo revealed that the 3V wall is not static and is amenable to cell movements. Furthermore, normally β-tanycytes give rise to parenchymal cells via an intermediate population of α-tanycytes with transient amplifying cell characteristics. Loss of Fgf10 temporarily attenuates the amplification of β-tanycytes but also appears to delay the exit of their α-tanycyte descendants from the germinal 3V wall. Our findings suggest that transience of cells through the α-tanycyte domain is a key feature, and Fgf10 is a negative regulator of postnatal hypothalamic neurogenesis. Summary: Generation of new hypothalamic neurons after birth is a multistep process involving cell division and cell movements that are controlled by Fgf10.
Collapse
Affiliation(s)
- Timothy Goodman
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Stuart G Nayar
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Shaun Clare
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Marta Mikolajczak
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, UK
| | - Ritva Rice
- Institute of Biotechnology, University of Helsinki, Viikinkaari 9, Helsinki 00014, Finland
| | - Suzanne Mansour
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112-5330, USA
| | - Saverio Bellusci
- Pediatrics, Saban Research Institute of Children's Hospital Los Angeles, University of California, Los Angeles, CA 90027, USA.,Excellence Cluster Cardio Pulmonary System, University Justus Liebig, 35392 Giessen, Germany
| | | |
Collapse
|
43
|
Vaid S, Huttner WB. Transcriptional Regulators and Human-Specific/Primate-Specific Genes in Neocortical Neurogenesis. Int J Mol Sci 2020; 21:ijms21134614. [PMID: 32610533 PMCID: PMC7369782 DOI: 10.3390/ijms21134614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
During development, starting from a pool of pluripotent stem cells, tissue-specific genetic programs help to shape and develop functional organs. To understand the development of an organ and its disorders, it is important to understand the spatio-temporal dynamics of the gene expression profiles that occur during its development. Modifications in existing genes, the de-novo appearance of new genes, or, occasionally, even the loss of genes, can greatly affect the gene expression profile of any given tissue and contribute to the evolution of organs or of parts of organs. The neocortex is evolutionarily the most recent part of the brain, it is unique to mammals, and is the seat of our higher cognitive abilities. Progenitors that give rise to this tissue undergo sequential waves of differentiation to produce the complete sets of neurons and glial cells that make up a functional neocortex. We will review herein our understanding of the transcriptional regulators that control the neural precursor cells (NPCs) during the generation of the most abundant class of neocortical neurons, the glutametergic neurons. In addition, we will discuss the roles of recently-identified human- and primate-specific genes in promoting neurogenesis, leading to neocortical expansion.
Collapse
|
44
|
Bertacchi M, Romano AL, Loubat A, Tran Mau-Them F, Willems M, Faivre L, Khau van Kien P, Perrin L, Devillard F, Sorlin A, Kuentz P, Philippe C, Garde A, Neri F, Di Giaimo R, Oliviero S, Cappello S, D'Incerti L, Frassoni C, Studer M. NR2F1 regulates regional progenitor dynamics in the mouse neocortex and cortical gyrification in BBSOAS patients. EMBO J 2020; 39:e104163. [PMID: 32484994 PMCID: PMC7327499 DOI: 10.15252/embj.2019104163] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/01/2020] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
The relationships between impaired cortical development and consequent malformations in neurodevelopmental disorders, as well as the genes implicated in these processes, are not fully elucidated to date. In this study, we report six novel cases of patients affected by BBSOAS (Boonstra‐Bosch‐Schaff optic atrophy syndrome), a newly emerging rare neurodevelopmental disorder, caused by loss‐of‐function mutations of the transcriptional regulator NR2F1. Young patients with NR2F1 haploinsufficiency display mild to moderate intellectual disability and show reproducible polymicrogyria‐like brain malformations in the parietal and occipital cortex. Using a recently established BBSOAS mouse model, we found that Nr2f1 regionally controls long‐term self‐renewal of neural progenitor cells via modulation of cell cycle genes and key cortical development master genes, such as Pax6. In the human fetal cortex, distinct NR2F1 expression levels encompass gyri and sulci and correlate with local degrees of neurogenic activity. In addition, reduced NR2F1 levels in cerebral organoids affect neurogenesis and PAX6 expression. We propose NR2F1 as an area‐specific regulator of mouse and human brain morphology and a novel causative gene of abnormal gyrification.
Collapse
Affiliation(s)
- Michele Bertacchi
- Université Côte d'Azur, CNRS, Inserm, iBV, Paris, France.,Clinical and Experimental Epileptology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | | | - Agnès Loubat
- Université Côte d'Azur, CNRS, Inserm, iBV, Paris, France
| | - Frederic Tran Mau-Them
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Marjolaine Willems
- Hôpital Arnaud de Villeneuve, Service de Génétique Médicale, CHU de Montpellier, Montpellier, France
| | - Laurence Faivre
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de Référence maladies rares « Anomalies du développement et syndromes malformatifs », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Philippe Khau van Kien
- Hôpital Carémeau, UF de Génétique Médicale et Cytogénétique, Centre de Compétences Anomalies du Développement et Syndromes Malformatifs, CHU de Nîmes, Nîmes, France
| | - Laurence Perrin
- Unité Fonctionnelle de Génétique Clinique, Hôpital Robert Debré, Paris, France
| | - Françoise Devillard
- Département de Génétique et Procréation, Hôpital Couple-Enfant, CHU de Grenoble, Grenoble, France
| | - Arthur Sorlin
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de Référence maladies rares « Anomalies du développement et syndromes malformatifs », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de référence maladies rares « Déficiences intellectuelles de causes rares », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Paul Kuentz
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Génétique Biologique, PCBio, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Christophe Philippe
- UMR1231 GAD, Inserm - Université Bourgogne-Franche Comté, Dijon, France.,Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Aurore Garde
- Unité Fonctionnelle Innovation en Diagnostic Génomique des Maladies Rares, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France.,Centre de Référence maladies rares « Anomalies du développement et syndromes malformatifs », Centre de Génétique, FHU-TRANSLAD, CHU Dijon Bourgogne, Dijon, France
| | - Francesco Neri
- Epigenetics Unit, Italian Institute for Genomic Medicine, University of Torino, Torino, Italy.,Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, Napoli, Italy.,Max Planck Institute of Psychiatry, München, Germany
| | - Salvatore Oliviero
- Epigenetics Unit, Italian Institute for Genomic Medicine, University of Torino, Torino, Italy
| | | | - Ludovico D'Incerti
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Carolina Frassoni
- Clinical and Experimental Epileptology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, iBV, Paris, France
| |
Collapse
|
45
|
Matsumoto N, Tanaka S, Horiike T, Shinmyo Y, Kawasaki H. A discrete subtype of neural progenitor crucial for cortical folding in the gyrencephalic mammalian brain. eLife 2020; 9:54873. [PMID: 32312384 PMCID: PMC7173966 DOI: 10.7554/elife.54873] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/01/2020] [Indexed: 12/28/2022] Open
Abstract
An increase in the diversity of neural progenitor subtypes and folding of the cerebral cortex are characteristic features which appeared during the evolution of the mammalian brain. Here, we show that the expansion of a specific subtype of neural progenitor is crucial for cortical folding. We found that outer radial glial (oRG) cells can be subdivided by HOPX expression in the gyrencephalic cerebral cortex of ferrets. Compared with HOPX-negative oRG cells, HOPX-positive oRG cells had high self-renewal activity and were accumulated in prospective gyral regions. Using our in vivo genetic manipulation technique for ferrets, we found that the number of HOPX-positive oRG cells and their self-renewal activity were regulated by sonic hedgehog (Shh) signaling. Importantly, suppressing Shh signaling reduced HOPX-positive oRG cells and cortical folding, while enhancing it had opposing effects. Our results reveal a novel subtype of neural progenitor important for cortical folding in gyrencephalic mammalian cerebral cortex.
Collapse
Affiliation(s)
- Naoyuki Matsumoto
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Satoshi Tanaka
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.,Medical Research Training Program, School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Toshihide Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
46
|
Cárdenas A, Borrell V. Molecular and cellular evolution of corticogenesis in amniotes. Cell Mol Life Sci 2020; 77:1435-1460. [PMID: 31563997 PMCID: PMC11104948 DOI: 10.1007/s00018-019-03315-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 09/03/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
The cerebral cortex varies dramatically in size and complexity between amniotes due to differences in neuron number and composition. These differences emerge during embryonic development as a result of variations in neurogenesis, which are thought to recapitulate modifications occurred during evolution that culminated in the human neocortex. Here, we review work from the last few decades leading to our current understanding of the evolution of neurogenesis and size of the cerebral cortex. Focused on specific examples across vertebrate and amniote phylogeny, we discuss developmental mechanisms regulating the emergence, lineage, complexification and fate of cortical germinal layers and progenitor cell types. At the cellular level, we discuss the fundamental impact of basal progenitor cells and the advent of indirect neurogenesis on the increased number and diversity of cortical neurons and layers in mammals, and on cortex folding. Finally, we discuss recent work that unveils genetic and molecular mechanisms underlying this progressive expansion and increased complexity of the amniote cerebral cortex during evolution, with a particular focus on those leading to human-specific features. Whereas new genes important in human brain development emerged the recent hominid lineage, regulation of the patterns and levels of activity of highly conserved signaling pathways are beginning to emerge as mechanisms of central importance in the evolutionary increase in cortical size and complexity across amniotes.
Collapse
Affiliation(s)
- Adrián Cárdenas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernández, 03550, Sant Joan d'Alacant, Alicante, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas y Universidad Miguel Hernández, 03550, Sant Joan d'Alacant, Alicante, Spain.
| |
Collapse
|
47
|
Güven A, Kalebic N, Long KR, Florio M, Vaid S, Brandl H, Stenzel D, Huttner WB. Extracellular matrix-inducing Sox9 promotes both basal progenitor proliferation and gliogenesis in developing neocortex. eLife 2020; 9:49808. [PMID: 32191207 PMCID: PMC7105383 DOI: 10.7554/elife.49808] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Neocortex expansion is largely based on the proliferative capacity of basal progenitors (BPs), which is increased by extracellular matrix (ECM) components via integrin signaling. Here we show that the transcription factor Sox9 drives expression of ECM components and that laminin 211 increases BP proliferation in embryonic mouse neocortex. We show that Sox9 is expressed in human and ferret BPs and is required for BP proliferation in embryonic ferret neocortex. Conditional Sox9 expression in the mouse BP lineage, where it normally is not expressed, increases BP proliferation, reduces Tbr2 levels and induces Olig2 expression, indicative of premature gliogenesis. Conditional Sox9 expression also results in cell-non-autonomous stimulation of BP proliferation followed by increased upper-layer neuron production. Our findings demonstrate that Sox9 exerts concerted effects on transcription, BP proliferation, neuron production, and neurogenic vs. gliogenic BP cell fate, suggesting that Sox9 may have contributed to promote neocortical expansion.
Collapse
Affiliation(s)
- Ayse Güven
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Human Technopole, Milan, Italy
| | - Katherine R Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marta Florio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Holger Brandl
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Denise Stenzel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
48
|
Namba T, Dóczi J, Pinson A, Xing L, Kalebic N, Wilsch-Bräuninger M, Long KR, Vaid S, Lauer J, Bogdanova A, Borgonovo B, Shevchenko A, Keller P, Drechsel D, Kurzchalia T, Wimberger P, Chinopoulos C, Huttner WB. Human-Specific ARHGAP11B Acts in Mitochondria to Expand Neocortical Progenitors by Glutaminolysis. Neuron 2020; 105:867-881.e9. [PMID: 31883789 DOI: 10.1016/j.neuron.2019.11.027] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/28/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022]
Abstract
The human-specific gene ARHGAP11B is preferentially expressed in neural progenitors of fetal human neocortex and increases abundance and proliferation of basal progenitors (BPs), which have a key role in neocortex expansion. ARHGAP11B has therefore been implicated in the evolutionary expansion of the human neocortex, but its mode of action has been unknown. Here, we show that ARHGAP11B is imported into mitochondria, where it interacts with the adenine nucleotide translocase (ANT) and inhibits the mitochondrial permeability transition pore (mPTP). BP expansion by ARHGAP11B requires its presence in mitochondria, and pharmacological inhibition of ANT function or mPTP opening mimic BP expansion by ARHGAP11B. Searching for the underlying metabolic basis, we find that BP expansion by ARHGAP11B requires glutaminolysis, the conversion of glutamine to glutamate for the tricarboxylic acid (TCA) cycle. Hence, an ARHGAP11B-induced, mitochondria-based effect on BP metabolism that is a hallmark of highly mitotically active cells appears to underlie its role in neocortex expansion.
Collapse
Affiliation(s)
- Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany.
| | - Judit Dóczi
- Department of Medical Biochemistry, Semmelweis University, Budapest, Tuzolto St. 37-47 1094, Hungary
| | - Anneline Pinson
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Michaela Wilsch-Bräuninger
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Katherine R Long
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Janelle Lauer
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Aliona Bogdanova
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Barbara Borgonovo
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Anna Shevchenko
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Patrick Keller
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - David Drechsel
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Teymuras Kurzchalia
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Pauline Wimberger
- Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Technische Universität Dresden, Dresden, Germany
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, Tuzolto St. 37-47 1094, Hungary
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany.
| |
Collapse
|
49
|
Cell stress in cortical organoids impairs molecular subtype specification. Nature 2020; 578:142-148. [PMID: 31996853 PMCID: PMC7433012 DOI: 10.1038/s41586-020-1962-0] [Citation(s) in RCA: 349] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 11/26/2019] [Indexed: 01/09/2023]
Abstract
Cortical organoids are self-organizing three-dimensional cultures that model features of the developing human cerebral cortex1,2. However, the fidelity of organoid models remains unclear3-5. Here we analyse the transcriptomes of individual primary human cortical cells from different developmental periods and cortical areas. We find that cortical development is characterized by progenitor maturation trajectories, the emergence of diverse cell subtypes and areal specification of newborn neurons. By contrast, organoids contain broad cell classes, but do not recapitulate distinct cellular subtype identities and appropriate progenitor maturation. Although the molecular signatures of cortical areas emerge in organoid neurons, they are not spatially segregated. Organoids also ectopically activate cellular stress pathways, which impairs cell-type specification. However, organoid stress and subtype defects are alleviated by transplantation into the mouse cortex. Together, these datasets and analytical tools provide a framework for evaluating and improving the accuracy of cortical organoids as models of human brain development.
Collapse
|
50
|
Lee JH, Shaker MR, Lee E, Lee B, Sun W. NeuroCore formation during differentiation of neurospheres of mouse embryonic neural stem cells. Stem Cell Res 2020; 43:101691. [PMID: 32018208 DOI: 10.1016/j.scr.2019.101691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 11/18/2022] Open
Abstract
Neural stem cells (NSCs) in the embryonic neocortex have the potential to generate a well-organized laminar architecture of the cerebral cortex through precise regulation of the proliferation, differentiation, and migration of neural cells. NSCs can be isolated in vitro and expanded as cell clusters, called neurospheres, which are primarily related to the proliferation ability of NSCs. Conversely, the tissue-organizing properties of NSCs via regulated differentiation and migration of the cells are not well understood. In this study, we established a three-dimensional (3D) differentiation model of neurospheres, which produce unique neuronal clusters, termed NeuroCore (NC). NC formation was initiated by the aggregation of young neurons. Upon maturation of the neurons and the establishment of radial glia-like structures, the initial organization of the NCs transformed into a glomeruli-like arrangement of cortical neurons. These neurons expressed multiple markers of upper and deep cortical neurons. Taken together, we propose that NSCs in vitro maintain some aspects of their original in vivo tissue-organizing properties, providing an alternative opportunity to explore the fundamental components of brain histogenesis in vitro.
Collapse
Affiliation(s)
- Ju-Hyun Lee
- Department of Anatomy and Division of Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Mohammed R Shaker
- Department of Anatomy and Division of Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Eunsoo Lee
- Department of Anatomy and Division of Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Boram Lee
- Department of Anatomy and Division of Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy and Division of Brain Korea 21 Plus Program for Biomedical Science, Korea University College of Medicine, 73, Inchon-ro, Seongbuk-gu, Seoul 02841, Republic of Korea.
| |
Collapse
|