1
|
Abusaada A, De Rosa F, Luhmann HJ, Kilb W, Sinning A. GABAergic integration of transient and persistent neurons in the developing mouse somatosensory cortex. Front Cell Neurosci 2025; 19:1556174. [PMID: 40078325 PMCID: PMC11897519 DOI: 10.3389/fncel.2025.1556174] [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: 01/06/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
GABA is an essential element in the function of neocortical circuits. The origin, migration and mechanisms of synaptogenesis of GABAergic neurons have been intensively studied. However, little information is available when GABAergic synapses are formed within the different cortical layers, neuronal cell types and subcellular compartments. To quantify the distribution of GABAergic synapses in the immature somatosensory mouse cortex, GABAergic synapses were identified by spatially coincident immunoprofiles for the pre- and postsynaptic markers vGAT and gephyrin at postnatal days (P)0-12. Between P0-5, GABAergic synapses are mainly restricted to the marginal zone, while at later developmental stages a more homogenous distribution is obtained. Cajal-Retzius neurons represent a major target of GABAergic synapses in the marginal zone with a homogeneous synapse distribution along the dendrite. The number of GABAergic synapses per pyramidal neuron increases substantially between P0 and P12, with a stable density and distribution in basal dendrites. In contrast, along apical dendrites synapses accumulate to more proximal positions after P8. Overall, the results of this study demonstrate that early GABAergic synaptogenesis is characterized by a consistent increase in the density of synapses with first a stringent overrepresentation in the marginal zone and a delayed establishment of perisomatic synapses in pyramidal neurons.
Collapse
|
2
|
Qiu H, Zhang M, Chen C, Wang H, Yue X. Decreasing β-Catenin Leads to Altered Endothelial Morphology, Increased Barrier Permeability and Cognitive Impairment During Chronic Methamphetamine Exposure. Int J Mol Sci 2025; 26:1514. [PMID: 40003980 PMCID: PMC11854931 DOI: 10.3390/ijms26041514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Cognitive impairment induced by chronic methamphetamine (METH) exposure exhibits similarities to neurodegenerative disorders and is associated with blood-brain barrier (BBB) dysfunction. However, the potential involvement of β-catenin in maintaining BBB integrity during METH exposure remains unexplored. In this study, Y-maze and novel object recognition tests were conducted to assess cognitive impairment in mice exposed chronically to methamphetamine for 2 and 4 weeks. Gd-DTPA and Evans blue leakage tests revealed disruption of the BBB in the hippocampus, while chronic METH exposure for 2 and 4 weeks significantly decreased β-catenin levels along with its transcriptionally regulated protein, claudin5. Additionally, various neural injury-related proteins, such as APP, Aβ1-42, p-tau (Thr181) and p-tau (Ser396), as well as neuroinflammation-related proteins, such as IL-6, IL-1β, and TNF-α, exhibited increased levels following chronic METH exposure. Furthermore, plasma analysis indicated elevated levels of p-Tau (total), neurofilament light chain, and GFAP. In vitro experiments demonstrated that exposure to METH resulted in dose-dependent and time-dependent reductions in cellular activity and connectivity of bEnd.3 and hcmec/D3 cells. Furthermore, β-catenin exhibited decreased levels and altered subcellular localization, transitioning from the cell membrane to the cytoplasm and nucleus upon METH exposure. Overexpression of β-catenin was found to alleviate endothelial toxicity and attenuate junctional weakening induced by METH. The aforementioned findings underscore the crucial involvement of β-catenin in endothelial cells during chronic METH exposure-induced disruption of the BBB, thereby presenting a potential novel target for addressing METH-associated cerebrovascular dysfunction and cognitive impairment.
Collapse
Affiliation(s)
| | | | | | - Huijun Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; (H.Q.); (M.Z.); (C.C.)
| | - Xia Yue
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China; (H.Q.); (M.Z.); (C.C.)
| |
Collapse
|
3
|
Zanetti A, Dujardin G, Fares-Taie L, Amiel J, Roger JE, Audo I, Robert MP, David P, Jung V, Goudin N, Guerrera IC, Moriceau S, Amana D, Assia Batzir N, Bachar-Zipori A, Basel Salmon L, Boddaert N, Briault S, Bruel AL, Costet-Fighiera C, Coutinho Santos L, Gitiaux C, Kaminska K, Kuentz P, Orenstein N, Philip-Sarles N, Plutino M, Quinodoz M, Santos C, Sigaudy S, Soeiro E Sá M, Sofrin E, Sousa AB, Sousa-Luis R, Thauvin-Robinet C, van Dijk EL, Zaafrane-Khachnaoui K, Zur D, Kaplan J, Rivolta C, Rozet JM, Perrault I. GPATCH11 variants cause mis-splicing and early-onset retinal dystrophy with neurological impairment. Nat Commun 2024; 15:10096. [PMID: 39572588 PMCID: PMC11582697 DOI: 10.1038/s41467-024-54549-8] [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: 10/09/2023] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
Here we conduct a study involving 12 individuals with retinal dystrophy, neurological impairment, and skeletal abnormalities, with special focus on GPATCH11, a lesser-known G-patch domain-containing protein, regulator of RNA metabolism. To elucidate its role, we study fibroblasts from unaffected individuals and patients carrying the recurring c.328+1 G > T mutation, which specifically removes the main part of the G-patch domain while preserving the other domains. Additionally, we generate a mouse model replicating the patients' phenotypic defects, including retinal dystrophy and behavioral abnormalities. Our results reveal a subcellular localization of GPATCH11 characterized by a diffuse presence in the nucleoplasm, as well as centrosomal localization, suggesting potential functions in RNA and cilia metabolism. Transcriptomic analysis performed on mouse retina detect dysregulation in both gene expression and splicing activity, impacting key processes such as photoreceptor light responses, RNA regulation, and primary cilia-associated metabolism. Proteomic analysis of mouse retina confirms the roles GPATCH11 plays in RNA processing, splicing, and transcription regulation, while also suggesting additional functions in synaptic plasticity and nuclear stress response. Our research provides insights into the diverse roles of GPATCH11 and identifies that the mutations affecting this protein are responsible for a recently characterized described syndrome.
Collapse
Affiliation(s)
- Andrea Zanetti
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Cité University, Paris, France
| | - Gwendal Dujardin
- Génétique, Génomique fonctionnelle et Biotechnologies (GGB), Université de Brest, INSERM UMR1078, EFS, Brest, France
| | - Lucas Fares-Taie
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Cité University, Paris, France
| | - Jeanne Amiel
- Laboratory of Embryology and Genetics of Malformations, INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Cité University, Paris, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neurosciences, CERTO-Retina France, CNRS, Paris-Saclay University, Saclay, France
| | - Isabelle Audo
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, National Rare Disease Center REFERET F-, Paris, France
| | - Matthieu P Robert
- Ophthalmology Department, University Hospital Necker-Enfants Malades, APHP, Paris, France
| | - Pierre David
- Transgenesis platform, Laboratory of Animal Experimentation and Transgenesis (LEAT) of the Structure Fédérative de Recherche Necker, INSERM US24/CNRS UMSS3633, Institute of Genetic Diseases, Imagine, Paris, France
| | - Vincent Jung
- Proteomic Platform Necker, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Nicolas Goudin
- Necker Bioimage Analysis Core Facility of the Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Ida Chiara Guerrera
- Proteomic Platform Necker, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Stéphanie Moriceau
- Platform for Neurobehavioral and metabolism, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Institute of Genetic Diseases, Imagine, Paris, France
| | - Danielle Amana
- Ophthalmology Department, Hospital Center of Orleans, Orleans, France
| | - Nurit Assia Batzir
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Anat Bachar-Zipori
- Ophthalmology Division, Tel Aviv Medical Center; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Lina Basel Salmon
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Recanati Genetics Institute, Rabin Medical Center, Petah Tikva, Israel
- Felsenstein Medical Research Center, Petah Tikva, Israel
| | - Nathalie Boddaert
- Pediatric-Radiology Department, University Hospital Necker-Enfants Malades, APHP, Paris Cité University, INSERM UMR1163, Paris, France
| | - Sylvain Briault
- Genetics Department, Regional Hospital of Orleans (CHRO), Orleans, France
| | - Ange-Line Bruel
- INSERM UMR1231, GAD team Université de Bourgogne-Franche Comté, FHU-TRANSLAD, CHU Dijon, Dijon, France
| | | | | | - Cyril Gitiaux
- Department of Clinical Neurophysiology, Reference center for neuromuscular pathologies Paris Nord Est, University Hospital Necker-Enfants Malades, Paris Cité University, Paris, France
| | - Karolina Kaminska
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Paul Kuentz
- INSERM UMR1231, GAD team Université de Bourgogne-Franche Comté, FHU-TRANSLAD, CHU Dijon, Dijon, France
| | - Naama Orenstein
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Morgane Plutino
- Service de Génétique Médicale, Hôpital l'Archet 2, CHU de Nice, Nice, France
| | - Mathieu Quinodoz
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Cristina Santos
- Instituto de Oftalmologia Dr. Gama Pinto (IOGP), Lisboa, Portugal
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Sabine Sigaudy
- Medical Genetics Department, Hospital Timone Enfant, Marseille, France
| | - Mariana Soeiro E Sá
- Department of Medical Genetics, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Efrat Sofrin
- Pediatric Genetics Unit, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Ana Berta Sousa
- Department of Medical Genetics, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Laboratory of Basic Immunology, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Rui Sousa-Luis
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Christel Thauvin-Robinet
- INSERM UMR1231, GAD team Université de Bourgogne-Franche Comté, FHU-TRANSLAD, CHU Dijon, Dijon, France
- Reference Center for Rare Diseases "Developmental Abnormalities and Malformation Syndromes" of the East, Genetic center, Hopital d'Enfants, FHU TRANSLAD, CHU Dijon, Dijon, France
| | - Erwin L van Dijk
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette Cedex, France
| | | | - Dinah Zur
- Ophthalmology Division, Tel Aviv Medical Center; Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Josseline Kaplan
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Cité University, Paris, France
| | - Carlo Rivolta
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Jean-Michel Rozet
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Cité University, Paris, France
| | - Isabelle Perrault
- Laboratory of Genetics in Ophthalmology (LGO), INSERM UMR1163, Institute of Genetic Diseases, Imagine and Paris Cité University, Paris, France.
| |
Collapse
|
4
|
Liu KE, Kucenas S. Rohon-beard neurons do not succumb to programmed cell death during zebrafish development. Dev Biol 2024; 515:186-198. [PMID: 38944329 DOI: 10.1016/j.ydbio.2024.06.020] [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: 02/12/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
During neural development, sculpting of early formed circuits by cell death and synaptic pruning is necessary to generate a functional and efficient nervous system. This allows for the establishment of rudimentary circuits which necessitate early organism survival to later undergo subsequent refinement. These changes facilitate additional specificity to stimuli which can lead to increased behavioral complexity. In multiple species, Rohon-Beard neurons (RBs) are the earliest mechanosensory neurons specified and are critical in establishing a rudimentary motor response circuit. Sensory input from RBs gradually becomes redundant as dorsal root ganglion (DRG) neurons develop and integrate into motor circuits. Previous studies demonstrate that RBs undergo a dramatic wave of cell death concurrent with development of the DRG. However, contrary to these studies, we show that neurogenin1+ (ngn1) RBs do not undergo a widespread wave of programmed cell death during early zebrafish development and instead persist until at least 15 days post fertilization (dpf). Starting at 2 dpf, we also observed a dramatic medialization and shrinkage of ngn1+ RB somas along with a gradual downregulation of ngn1 in RBs. This alters a fundamental premise of early zebrafish neural development and opens new avenues to explore mechanisms of RB function, persistence, and circuit refinement.
Collapse
Affiliation(s)
- Kendra E Liu
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, 22904, USA; Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA
| | - Sarah Kucenas
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, 22904, USA; Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA; Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, 22904, USA.
| |
Collapse
|
5
|
Méndez P, de la Vega-Ruiz R, Montes-Mellado A. Estrogenic regulation of hippocampal inhibitory system across lifespan. J Neuroendocrinol 2024:e13441. [PMID: 39143852 DOI: 10.1111/jne.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Estrogens produced in peripheral tissues and locally in the brain are potent neuromodulators. The function of the hippocampus, a brain region essential for episodic memory and spatial navigation, relies on the activity of ensembles of excitatory neurons whose activity is temporally and spatially coordinated by a wide diversity of inhibitory neurons (INs) types. Over the last years, we have accumulated evidence that indicates that estrogens regulate the function of hippocampal INs through different mechanisms, including transcriptional regulation and rapid nongenomic signaling. Here, we argue that the well-documented influence of estrogens on episodic memory may be related to the actions of local and peripheral estrogens on the heterogenous populations of hippocampal INs. We discuss how physiological changes in peripheral sex hormone levels throughout lifespan may interact with local brain sources to regulate IN function at different stages of life, from early hippocampal development to the aging brain. We conclude that considering INs as mediators of sex hormone actions in the hippocampus across the healthy life span will benefit our understanding of sex-biased neurodevelopmental disorders and physiological aging.
Collapse
|
6
|
Glærum IL, Dunville K, Moan K, Krause M, Montaldo NP, Kirikae H, Nigro MJ, Sætrom P, van Loon B, Quattrocolo G. Postnatal persistence of hippocampal Cajal-Retzius cells has a crucial role in the establishment of the hippocampal circuit. Development 2024; 151:dev202236. [PMID: 38095282 PMCID: PMC10820737 DOI: 10.1242/dev.202236] [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: 08/04/2023] [Accepted: 11/27/2023] [Indexed: 01/10/2024]
Abstract
Cajal-Retzius (CR) cells are a transient neuron type that populate the postnatal hippocampus. To understand how the persistence of CR cells influences the maturation of hippocampal circuits, we combined a specific transgenic mouse line with viral vector injection to selectively ablate CR cells from the postnatal hippocampus. We observed layer-specific changes in the dendritic complexity and spine density of CA1 pyramidal cells. In addition, transcriptomic analysis highlighted significant changes in the expression of synapse-related genes across development. Finally, we were able to identify significant changes in the expression levels of latrophilin 2, a postsynaptic guidance molecule known for its role in the entorhinal-hippocampal connectivity. These findings were supported by changes in the synaptic proteomic content in CA1 stratum lacunosum-moleculare. Our results reveal a crucial role for CR cells in the establishment of the hippocampal network.
Collapse
Affiliation(s)
- Ingvild Lynneberg Glærum
- Kavli Institute for Systems Neuroscience and Center for Algorithms of the Cortex, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
- Mohn Research Center for the Brain, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Keagan Dunville
- Kavli Institute for Systems Neuroscience and Center for Algorithms of the Cortex, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Kristian Moan
- Kavli Institute for Systems Neuroscience and Center for Algorithms of the Cortex, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Maike Krause
- Kavli Institute for Systems Neuroscience and Center for Algorithms of the Cortex, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Nicola Pietro Montaldo
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Hinako Kirikae
- Kavli Institute for Systems Neuroscience and Center for Algorithms of the Cortex, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Maximiliano Jose Nigro
- Kavli Institute for Systems Neuroscience and Center for Algorithms of the Cortex, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Pål Sætrom
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Barbara van Loon
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| | - Giulia Quattrocolo
- Kavli Institute for Systems Neuroscience and Center for Algorithms of the Cortex, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
- Mohn Research Center for the Brain, Norwegian University of Science and Technology (NTNU), Trondheim 7491, Norway
| |
Collapse
|
7
|
van Bruggen R, Patel ZH, Wang M, Suk TR, Rousseaux MWC, Tan Q. A Versatile Strategy for Genetic Manipulation of Cajal-Retzius Cells in the Adult Mouse Hippocampus. eNeuro 2023; 10:ENEURO.0054-23.2023. [PMID: 37775311 PMCID: PMC10585607 DOI: 10.1523/eneuro.0054-23.2023] [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: 02/14/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023] Open
Abstract
Cajal-Retzius (CR) cells are transient neurons with long-lasting effects on the architecture and circuitry of the neocortex and hippocampus. Contrary to the prevailing assumption that CR cells completely disappear in rodents shortly after birth, a substantial portion of these cells persist in the hippocampus throughout adulthood. The role of these surviving CR cells in the adult hippocampus is largely unknown, partly because of the paucity of suitable tools to dissect their functions in the adult versus the embryonic brain. Here, we show that genetic crosses of the ΔNp73-Cre mouse line, widely used to target CR cells, to reporter mice induce reporter expression not only in CR cells, but also progressively in postnatal dentate gyrus granule neurons. Such a lack of specificity may confound studies of CR cell function in the adult hippocampus. To overcome this, we devise a method that not only leverages the temporary CR cell-targeting specificity of the ΔNp73-Cre mice before the first postnatal week, but also capitalizes on the simplicity and effectiveness of freehand neonatal intracerebroventricular injection of adeno-associated virus. We achieve robust Cre-mediated recombination that remains largely restricted to hippocampal CR cells from early postnatal age to adulthood. We further demonstrate the utility of this method to manipulate neuronal activity of CR cells in the adult hippocampus. This versatile and scalable strategy will facilitate experiments of CR cell-specific gene knockdown and/or overexpression, lineage tracing, and neural activity modulation in the postnatal and adult brain.
Collapse
Affiliation(s)
- Rebekah van Bruggen
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Zain H Patel
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Mi Wang
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Terry R Suk
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Eric Poulin Center for Neuromuscular Diseases, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Maxime W C Rousseaux
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Eric Poulin Center for Neuromuscular Diseases, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Qiumin Tan
- Department of Cell Biology, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta T6G 1C9, Canada
| |
Collapse
|
8
|
Enck JR, Olson EC. Calcium Signaling during Cortical Apical Dendrite Initiation: A Role for Cajal-Retzius Neurons. Int J Mol Sci 2023; 24:12965. [PMID: 37629145 PMCID: PMC10455361 DOI: 10.3390/ijms241612965] [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: 06/12/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The apical dendrite of a cortical projection neuron (CPN) is generated from the leading process of the migrating neuron as the neuron completes migration. This transformation occurs in the cortical marginal zone (MZ), a layer that contains the Cajal-Retzius neurons and their axonal projections. Cajal-Retzius neurons (CRNs) are well known for their critical role in secreting Reelin, a glycoprotein that controls dendritogenesis and cell positioning in many regions of the developing brain. In this study, we examine the possibility that CRNs in the MZ may provide additional signals to arriving CPNs, that may promote the maturation of CPNs and thus shape the development of the cortex. We use whole embryonic hemisphere explants and multiphoton microscopy to confirm that CRNs display intracellular calcium transients of <1-min duration and high amplitude during early corticogenesis. In contrast, developing CPNs do not show high-amplitude calcium transients, but instead show a steady increase in intracellular calcium that begins at the time of dendritic initiation, when the leading process of the migrating CPN is encountering the MZ. The possible existence of CRN to CPN communication was revealed by the application of veratridine, a sodium channel activator, which has been shown to preferentially stimulate more mature cells in the MZ at an early developmental time. Surprisingly, veratridine application also triggers large calcium transients in CPNs, which can be partially blocked by a cocktail of antagonists that block glutamate and glycine receptor activation. These findings outline a model in which CRN spontaneous activity triggers the release of glutamate and glycine, neurotransmitters that can trigger intracellular calcium elevations in CPNs. These elevations begin as CPNs initiate dendritogenesis and continue as waves in the post-migratory cells. Moreover, we show that the pharmacological blockade of glutamatergic signaling disrupts migration, while forced expression of a bacterial voltage-gated calcium channel (CavMr) in the migrating neurons promotes dendritic growth and migration arrest. The identification of CRN to CPN signaling during early development provides insight into the observation that many autism-linked genes encode synaptic proteins that, paradoxically, are expressed in the developing cortex well before the appearance of synapses and the establishment of functional circuits.
Collapse
Affiliation(s)
| | - Eric C. Olson
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, 505 Irving Ave., Syracuse, NY 13210, USA;
| |
Collapse
|
9
|
Moreau MX, Saillour Y, Elorriaga V, Bouloudi B, Delberghe E, Deutsch Guerrero T, Ochandorena-Saa A, Maeso-Alonso L, Marques MM, Marin MC, Spassky N, Pierani A, Causeret F. Repurposing of the multiciliation gene regulatory network in fate specification of Cajal-Retzius neurons. Dev Cell 2023; 58:1365-1382.e6. [PMID: 37321213 DOI: 10.1016/j.devcel.2023.05.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/06/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023]
Abstract
Cajal-Retzius cells (CRs) are key players in cerebral cortex development, and they display a unique transcriptomic identity. Here, we use scRNA-seq to reconstruct the differentiation trajectory of mouse hem-derived CRs, and we unravel the transient expression of a complete gene module previously known to control multiciliogenesis. However, CRs do not undergo centriole amplification or multiciliation. Upon deletion of Gmnc, the master regulator of multiciliogenesis, CRs are initially produced but fail to reach their normal identity resulting in their massive apoptosis. We further dissect the contribution of multiciliation effector genes and identify Trp73 as a key determinant. Finally, we use in utero electroporation to demonstrate that the intrinsic competence of hem progenitors as well as the heterochronic expression of Gmnc prevent centriole amplification in the CR lineage. Our work exemplifies how the co-option of a complete gene module, repurposed to control a distinct process, may contribute to the emergence of novel cell identities.
Collapse
Affiliation(s)
- Matthieu X Moreau
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Yoann Saillour
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Vicente Elorriaga
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Benoît Bouloudi
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Elodie Delberghe
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Tanya Deutsch Guerrero
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Amaia Ochandorena-Saa
- Université Paris Cité, Imagine-Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, 75015 Paris, France
| | - Laura Maeso-Alonso
- Instituto de Biomedicina, y Departamento de Biología Molecular, Universidad de León, 24071 Leon, Spain
| | - Margarita M Marques
- Instituto de Desarrollo Ganadero y Sanidad Animal, y Departamento de Producción Animal, Universidad de León, 24071 Leon, Spain
| | - Maria C Marin
- Instituto de Biomedicina, y Departamento de Biología Molecular, Universidad de León, 24071 Leon, Spain
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Département de Biologie, Ecole Normale Supérieure, CNRS, INSERM, Université PSL, 75005 Paris, France
| | - Alessandra Pierani
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Frédéric Causeret
- Université Paris Cité, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, 75015 Paris, France; Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France.
| |
Collapse
|