Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters










Publication year range
1.
Dev Cell ; 58(15): 1365-1382.e6, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37321213

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.


Subject(s)
Cerebral Cortex , Gene Regulatory Networks , Mice , Animals , Cerebral Cortex/metabolism , Neurons/metabolism , Cell Differentiation/physiology , Neurogenesis/genetics
2.
J Comp Neurol ; 531(12): 1229-1243, 2023 08.
Article in English | MEDLINE | ID: mdl-37125418

ABSTRACT

In vertebrates, the embryonic olfactory epithelium contains progenitors that will give rise to distinct classes of neurons, including olfactory sensory neurons (OSNs; involved in odor detection), vomeronasal sensory neurons (VSNs; responsible for pheromone sensing), and gonadotropin-releasing hormone (GnRH) neurons that control the hypothalamic-pituitary-gonadal axis. Currently, these three neuronal lineages are usually believed to emerge from uniform pools of progenitors. Here, we found that the homeodomain transcription factor Dbx1 is expressed by neurogenic progenitors in the developing and adult mouse olfactory epithelium. We demonstrate that Dbx1 itself is dispensable for neuronal fate specification and global organization of the olfactory sensory system. Using lineage tracing, we characterize the contribution of Dbx1 lineages to OSN, VSN, and GnRH neuron populations and reveal an unexpected degree of diversity. Furthermore, we demonstrate that Dbx1-expressing progenitors remain neurogenic in the absence of the proneural gene Ascl1. Our work therefore points to the existence of distinct neurogenic programs in Dbx1-derived and other olfactory lineages.


Subject(s)
Olfactory Mucosa , Olfactory Receptor Neurons , Mice , Animals , Olfactory Receptor Neurons/metabolism , Transcription Factors/genetics , Gene Expression Regulation , Gonadotropin-Releasing Hormone/metabolism , Homeodomain Proteins/genetics
3.
Cell ; 186(9): 1819-1821, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37116467

ABSTRACT

Metabolic changes are essential for neurodevelopmental processes. However, little is known about how and when neuronal metabolic remodeling occurs to promote functional circuits. In this issue of Cell, Knaus et al. demonstrate that a temporary perinatal shift in metabolites and lipids is crucial for cortical neurons' survival and wiring.


Subject(s)
Neurons , Cell Survival , Neuronal Plasticity/physiology , Neurons/physiology
4.
Nat Commun ; 14(1): 1531, 2023 03 18.
Article in English | MEDLINE | ID: mdl-36934089

ABSTRACT

Cajal-Retzius cells (CRs) are transient neurons, disappearing almost completely in the postnatal neocortex by programmed cell death (PCD), with a percentage surviving up to adulthood in the hippocampus. Here, we evaluate CR's role in the establishment of adult neuronal and cognitive function using a mouse model preventing Bax-dependent PCD. CRs abnormal survival resulted in impairment of hippocampus-dependent memory, associated in vivo with attenuated theta oscillations and enhanced gamma activity in the dorsal CA1. At the cellular level, we observed transient changes in the number of NPY+ cells and altered CA1 pyramidal cell spine density. At the synaptic level, these changes translated into enhanced inhibitory currents in hippocampal pyramidal cells. Finally, adult mutants displayed an increased susceptibility to lethal tonic-clonic seizures in a kainate model of epilepsy. Our data reveal that aberrant survival of a small proportion of postnatal hippocampal CRs results in cognitive deficits and epilepsy-prone phenotypes in adulthood.


Subject(s)
Hippocampus , Neurons , Hippocampus/physiology , Memory Disorders/genetics , Memory Disorders/metabolism , Neurons/metabolism , Pyramidal Cells/physiology , Seizures/genetics , Seizures/metabolism , Animals , Mice
5.
Int J Mol Sci ; 24(6)2023 Mar 11.
Article in English | MEDLINE | ID: mdl-36982451

ABSTRACT

Cajal-Retzius cells (CRs) are a class of transient neurons in the mammalian cortex that play a critical role in cortical development. Neocortical CRs undergo almost complete elimination in the first two postnatal weeks in rodents and the persistence of CRs during postnatal life has been detected in pathological conditions related to epilepsy. However, it is unclear whether their persistence is a cause or consequence of these diseases. To decipher the molecular mechanisms involved in CR death, we investigated the contribution of the PI3K/AKT/mTOR pathway as it plays a critical role in cell survival. We first showed that this pathway is less active in CRs after birth before massive cell death. We also explored the spatio-temporal activation of both AKT and mTOR pathways and reveal area-specific differences along both the rostro-caudal and medio-lateral axes. Next, using genetic approaches to maintain an active pathway in CRs, we found that the removal of either PTEN or TSC1, two negative regulators of the pathway, lead to differential CR survivals, with a stronger effect in the Pten model. Persistent cells in this latter mutant are still active. They express more Reelin and their persistence is associated with an increase in the duration of kainate-induced seizures in females. Altogether, we show that the decrease in PI3K/AKT/mTOR activity in CRs primes these cells to death by possibly repressing a survival pathway, with the mTORC1 branch contributing less to the phenotype.


Subject(s)
Kainic Acid , Proto-Oncogene Proteins c-akt , Animals , Female , Kainic Acid/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Seizures/chemically induced , Mammals/metabolism
6.
Curr Opin Neurobiol ; 79: 102686, 2023 04.
Article in English | MEDLINE | ID: mdl-36774666

ABSTRACT

Cajal-Retzius cells (CRs) are a transient neuronal type of the developing cerebral cortex. Over the years, they have been shown or proposed to play important functions in neocortical and hippocampal morphogenesis, circuit formation, brain evolution and human pathology. Because of their short lifespan, CRs have been pictured as a purely developmental cell type, whose production and active elimination are both required for correct brain development. In this review, we present some of the findings that allow us to better appreciate the identity and diversity of this very special cell type, and propose a unified definition of what should be considered a Cajal-Retzius cell, especially when working with non-mammalian species or organoids. In addition, we highlight a flurry of recent studies pointing to the importance of CRs in the assembly of functional and dysfunctional cortical networks.


Subject(s)
Cerebral Cortex , Neurons , Humans , Neurons/physiology , Hippocampus/physiology
7.
JCI Insight ; 6(16)2021 08 23.
Article in English | MEDLINE | ID: mdl-34237032

ABSTRACT

Congenital microcephaly (MCPH) is a neurodevelopmental disease associated with mutations in genes encoding proteins involved in centrosomal and chromosomal dynamics during mitosis. Detailed MCPH pathogenesis at the cellular level is still elusive, given the diversity of MCPH genes and lack of comparative in vivo studies. By generating a series of CRISPR/Cas9-mediated genetic KOs, we report here that - whereas defects in spindle pole proteins (ASPM, MCPH5) result in mild MCPH during development - lack of centrosome (CDK5RAP2, MCPH3) or centriole (CEP135, MCPH8) regulators induces delayed chromosome segregation and chromosomal instability in neural progenitors (NPs). Our mouse model of MCPH8 suggests that loss of CEP135 results in centriole duplication defects, TP53 activation, and cell death of NPs. Trp53 ablation in a Cep135-deficient background prevents cell death but not MCPH, and it leads to subcortical heterotopias, a malformation seen in MCPH8 patients. These results suggest that MCPH in some MCPH patients can arise from the lack of adaptation to centriole defects in NPs and may lead to architectural defects if chromosomally unstable cells are not eliminated during brain development.


Subject(s)
Centrioles/genetics , Chromosomal Instability , Microcephaly/genetics , Neural Stem Cells/pathology , Animals , Brain/cytology , Brain/pathology , CRISPR-Cas Systems/genetics , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrioles/pathology , Disease Models, Animal , Embryo, Mammalian , Female , Humans , Male , Mice , Mice, Knockout , Microcephaly/pathology , Microscopy, Electron, Transmission , Molecular Imaging , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/ultrastructure , Primary Cell Culture , Time-Lapse Imaging , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Development ; 148(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34170322

ABSTRACT

In the developing cerebral cortex, how progenitors that seemingly display limited diversity end up producing a vast array of neurons remains a puzzling question. The prevailing model suggests that temporal maturation of progenitors is a key driver in the diversification of the neuronal output. However, temporal constraints are unlikely to account for all diversity, especially in the ventral and lateral pallium where neuronal types significantly differ from their dorsal neocortical counterparts born at the same time. In this study, we implemented single-cell RNAseq to sample the diversity of progenitors and neurons along the dorso-ventral axis of the early developing pallium. We first identified neuronal types, mapped them on the tissue and determined their origin through genetic tracing. We characterised progenitor diversity and disentangled the gene modules underlying temporal versus spatial regulations of neuronal specification. Finally, we reconstructed the developmental trajectories followed by ventral and dorsal pallial neurons to identify lineage-specific gene waves. Our data suggest a model by which discrete neuronal fate acquisition from a continuous gradient of progenitors results from the superimposition of spatial information and temporal maturation.


Subject(s)
Cerebral Cortex/metabolism , Neurons/metabolism , Transcriptome , Animals , Cell Differentiation/physiology , Cerebral Cortex/pathology , Embryo, Mammalian , Female , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins , Neurogenesis/physiology , Proto-Oncogene Proteins/metabolism
9.
Semin Cell Dev Biol ; 118: 35-49, 2021 10.
Article in English | MEDLINE | ID: mdl-34034988

ABSTRACT

A hierarchical development of cortical areas was suggested over a century ago, but the diversity and complexity of cortical hierarchy properties have so far prevented a formal demonstration. The aim of this review is to clarify the similarities and differences in the developmental processes underlying cortical development of primary and higher-order areas. We start by recapitulating the historical and recent advances underlying the biological principle of cortical hierarchy in adults. We then revisit the arguments for a hierarchical maturation of cortical areas, and further integrate the principles of cortical areas specification during embryonic and postnatal development. We highlight how the dramatic expansion in cortical size might have contributed to the increased number of association areas sustaining cognitive complexification in evolution. Finally, we summarize the recent observations of an alteration of cortical hierarchy in neuropsychiatric disorders and discuss their potential developmental origins.


Subject(s)
Cerebral Cortex/growth & development , Animals , Humans , Spatio-Temporal Analysis
10.
Development ; 148(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34047341

ABSTRACT

Cajal-Retzius neurons (CRs) are among the first-born neurons in the developing cortex of reptiles, birds and mammals, including humans. The peculiarity of CRs lies in the fact they are initially embedded into the immature neuronal network before being almost completely eliminated by cell death at the end of cortical development. CRs are best known for controlling the migration of glutamatergic neurons and the formation of cortical layers through the secretion of the glycoprotein reelin. However, they have been shown to play numerous additional key roles at many steps of cortical development, spanning from patterning and sizing functional areas to synaptogenesis. The use of genetic lineage tracing has allowed the discovery of their multiple ontogenetic origins, migratory routes, expression of molecular markers and death dynamics. Nowadays, single-cell technologies enable us to appreciate the molecular heterogeneity of CRs with an unprecedented resolution. In this Review, we discuss the morphological, electrophysiological, molecular and genetic criteria allowing the identification of CRs. We further expose the various sources, migration trajectories, developmental functions and death dynamics of CRs. Finally, we demonstrate how the analysis of public transcriptomic datasets allows extraction of the molecular signature of CRs throughout their transient life and consider their heterogeneity within and across species.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Cell Adhesion Molecules, Neuronal , Cell Death , Cerebral Cortex/growth & development , Extracellular Matrix Proteins , Hippocampus/growth & development , Humans , Nerve Tissue Proteins/genetics , Neurogenesis/physiology , Neurons/cytology , Reelin Protein , Serine Endopeptidases , Transcriptome
11.
Sci Rep ; 11(1): 8349, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863910

ABSTRACT

The piriform cortex (PC) is a major cortical processing center for the sense of smell that receives direct inputs from the olfactory bulb. In mice, the PC consists of three neuronal layers, which are populated by cells with distinct developmental origins. One origin of PC neurons is the pool of Dbx1-expressing neural progenitors located in the ventral pallium at the pallial-subpallial boundary. Since the precise mechanisms of PC neuron development are largely unknown, we sought to define the distribution, timing of neurogenesis, morphology and projection patterns of PC neurons from the Dbx1 lineage. We found that Dbx1-lineage neurons are preferentially distributed in layer 2 and enriched in the ventral portion of the PC. Further, Dbx1 neurons are early-born neurons and contribute to most neuronal subtypes in the PC. Our data also revealed an enrichment of Dbx1-lineage neurons in the ventral anterior PC that project to the orbitofrontal cortex. These findings suggest a specific association between the developmental origin of PC neurons and their neuronal properties.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , Neurons/physiology , Piriform Cortex/cytology , Piriform Cortex/physiology , Smell , Animals , Gene Expression , Mice, Knockout , Olfactory Bulb/physiology , Stem Cells/metabolism , Stem Cells/physiology
12.
Front Cell Dev Biol ; 8: 580657, 2020.
Article in English | MEDLINE | ID: mdl-33102486

ABSTRACT

During development the vast majority of cells that will later compose the mature cerebral cortex undergo extensive migration to reach their final position. In addition to intrinsically distinct migratory behaviors, cells encounter and respond to vastly different microenvironments. These range from axonal tracts to cell-dense matrices, electrically active regions and extracellular matrix components, which may all change overtime. Furthermore, migrating neurons themselves not only adapt to their microenvironment but also modify the local niche through cell-cell contacts, secreted factors and ions. In the radial dimension, the developing cortex is roughly divided into dense progenitor and cortical plate territories, and a less crowded intermediate zone. The cortical plate is bordered by the subplate and the marginal zone, which are populated by neurons with high electrical activity and characterized by sophisticated neuritic ramifications. Neuronal migration is influenced by these boundaries resulting in dramatic changes in migratory behaviors as well as morphology and electrical activity. Modifications in the levels of any of these parameters can lead to alterations and even arrest of migration. Recent work indicates that morphology and electrical activity of migrating neuron are interconnected and the aim of this review is to explore the extent of this connection. We will discuss on one hand how the response of migrating neurons is altered upon modification of their intrinsic electrical properties and whether, on the other hand, the electrical properties of the cellular environment can modify the morphology and electrical activity of migrating cortical neurons.

13.
Cell Rep ; 29(3): 645-658.e5, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618633

ABSTRACT

Changes in transcriptional regulation through cis-regulatory elements are thought to drive brain evolution. However, how this impacts the identity of primate cortical neurons is still unresolved. Here, we show that primate-specific cis-regulatory sequences upstream of the Dbx1 gene promote human-like expression in the mouse embryonic cerebral cortex, and this imparts cell identity. Indeed, while Dbx1 is expressed in highly restricted cortical progenitors in the mouse ventral pallium, it is maintained in neurons in primates. Phenocopy of the primate-like Dbx1 expression in mouse cortical progenitors induces ectopic Cajal-Retzius and subplate (SP) neurons, which are transient populations playing crucial roles in cortical development. A conditional expression solely in neurons uncouples mitotic and postmitotic activities of Dbx1 and exclusively promotes a SP-like fate. Our results highlight how transcriptional changes of a single fate determinant in postmitotic cells may contribute to the expansion of neuronal diversity during cortical evolution.


Subject(s)
Biological Evolution , Cerebral Cortex/metabolism , Homeodomain Proteins/metabolism , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Embryo, Mammalian/metabolism , Female , Homeodomain Proteins/genetics , Humans , Macaca , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Pregnancy , T-Box Domain Proteins/metabolism
14.
Nat Commun ; 10(1): 4249, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31534164

ABSTRACT

The first wave of oligodendrocyte precursor cells (firstOPCs) and most GABAergic interneurons share common embryonic origins. Cortical firstOPCs are thought to be replaced by other OPC populations shortly after birth, maintaining a consistent OPC density and making postnatal interactions between firstOPCs and ontogenetically-related interneurons unlikely. Challenging these ideas, we show that a cortical firstOPC subpopulation survives and forms functional cell clusters with lineage-related interneurons. Favored by a common embryonic origin, these clusters display unexpected preferential synaptic connectivity and are anatomically maintained after firstOPCs differentiate into myelinating oligodendrocytes. While the concomitant rescue of interneurons and firstOPCs committed to die causes an exacerbated neuronal inhibition, it abolishes interneuron-firstOPC high synaptic connectivity. Further, the number of other oligodendroglia populations increases through a non-cell-autonomous mechanism, impacting myelination. These findings demonstrate unprecedented roles of interneuron and firstOPC apoptosis in regulating lineage-related cell interactions and the homeostatic oligodendroglia density.


Subject(s)
Apoptosis/physiology , Interneurons/metabolism , Neurogenesis/physiology , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Animals , Central Nervous System/cytology , Central Nervous System/embryology , Female , GABAergic Neurons/cytology , Homeodomain Proteins/metabolism , Interneurons/cytology , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Oligodendroglia/cytology
15.
Cell Death Discov ; 5: 91, 2019.
Article in English | MEDLINE | ID: mdl-31069116

ABSTRACT

In multicellular organisms, cell death pathways allow the removal of abnormal or unwanted cells. Their dysregulation can lead either to excessive elimination or to inappropriate cell survival. Evolutionary constraints ensure that such pathways are strictly regulated in order to restrain their activation to the appropriate context. We have previously shown that the transmembrane receptor Kremen1 behaves as a dependence receptor, triggering cell death unless bound to its ligand Dickkopf1. In this study, we reveal that Kremen1 apoptotic signaling requires homodimerization of the receptor. Dickkopf1 binding inhibits Kremen1 multimerization and alleviates cell death, whereas forced dimerization increases apoptotic signaling. Furthermore, we show that Kremen2, a paralog of Kremen1, which bears no intrinsic apoptotic activity, binds and competes with Kremen1. Consequently, Kremen2 is a very potent inhibitor of Kremen1-induced cell death. Kremen1 was proposed to act as a tumor suppressor, preventing cancer cell survival in a ligand-poor environment. We found that KREMEN2 expression is increased in a large majority of cancers, suggesting it may confer increased survival capacity. Consistently, low KREMEN2 expression is a good prognostic for patient survival in a variety of cancers.

16.
Elife ; 82019 12 31.
Article in English | MEDLINE | ID: mdl-31891351

ABSTRACT

Programmed cell death and early activity contribute to the emergence of functional cortical circuits. While most neuronal populations are scaled-down by death, some subpopulations are entirely eliminated, raising the question of the importance of such demise for cortical wiring. Here, we addressed this issue by focusing on Cajal-Retzius neurons (CRs), key players in cortical development that are eliminated in postnatal mice in part via Bax-dependent apoptosis. Using Bax-conditional mutants and CR hyperpolarization, we show that the survival of electrically active subsets of CRs triggers an increase in both dendrite complexity and spine density of upper layer pyramidal neurons, leading to an excitation/inhibition imbalance. The survival of these CRs is induced by hyperpolarization, highlighting an interplay between early activity and neuronal elimination. Taken together, our study reveals a novel activity-dependent programmed cell death process required for the removal of transient immature neurons and the proper wiring of functional cortical circuits.


Subject(s)
Apoptosis/genetics , Neurogenesis/genetics , Pyramidal Cells/metabolism , bcl-2-Associated X Protein/genetics , Animals , Animals, Newborn , Cell Polarity/genetics , Cerebral Cortex/metabolism , Electric Stimulation , Interstitial Cells of Cajal/metabolism , Mice , Mutant Proteins/genetics , Pyramidal Cells/pathology
17.
Curr Opin Neurobiol ; 53: 35-42, 2018 12.
Article in English | MEDLINE | ID: mdl-29738999

ABSTRACT

The mature cerebral cortex only contains a fraction of the cells that are generated during embryonic development. Indeed some neuronal populations are produced in excess and later subjected to partial elimination whereas others are almost completely removed during the first two postnatal weeks in mice. Although the identity of cells that disappear, the time course and mechanisms of their death are becoming reasonably well established, the meaning of producing supernumerary cells still remains elusive. In this review, we focus on recent data that shed a new light on the mechanisms involved in adjusting cell numbers and discuss the significance of refinement versus complete elimination of cell populations in the developing cortex.


Subject(s)
Cell Death/physiology , Cerebral Cortex/growth & development , Embryonic Development/physiology , Neurons/physiology , Animals , Cerebral Cortex/cytology , Humans , Neurons/cytology
18.
Dev Biol ; 432(1): 24-33, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28625870

ABSTRACT

Transcription factors are key orchestrators of the emergence of neuronal diversity within the developing spinal cord. As such, the two paralogous proteins Pax3 and Pax7 regulate the specification of progenitor cells within the intermediate neural tube, by defining a neat segregation between those fated to form motor circuits and those involved in the integration of sensory inputs. To attain insights into the molecular means by which they control this process, we have performed detailed phenotypic analyses of the intermediate spinal interneurons (IN), namely the dI6, V0D, V0VCG and V1 populations in compound null mutants for Pax3 and Pax7. This has revealed that the levels of Pax3/7 proteins determine both the dorso-ventral extent and the number of cells produced in each subpopulation; with increasing levels leading to the dorsalisation of their fate. Furthermore, thanks to the examination of mutants in which Pax3 transcriptional activity is skewed either towards repression or activation, we demonstrate that this cell diversification process is mainly dictated by Pax3/7 ability to repress gene expression. Consistently, we show that Pax3 and Pax7 inhibit the expression of Dbx1 and of its repressor Prdm12, fate determinants of the V0 and V1 interneurons, respectively. Notably, we provide evidence for the activity of several cis-regulatory modules of Dbx1 to be sensitive to Pax3 and Pax7 transcriptional activity levels. Altogether, our study provides insights into how the redundancy within a TF family, together with discrete dynamics of expression profiles of each member, are exploited to generate cellular diversity. Furthermore, our data supports the model whereby cell fate choices in the neural tube do not rely on binary decisions but rather on inhibition of multiple alternative fates.


Subject(s)
Homeodomain Proteins/physiology , Interneurons/physiology , Nerve Tissue Proteins/physiology , PAX3 Transcription Factor/physiology , PAX7 Transcription Factor/physiology , Spinal Cord/cytology , Animals , Cell Differentiation/physiology , Chick Embryo , Gene Expression Regulation, Developmental , Interneurons/cytology , Mice , Neural Tube/physiology , Spinal Cord/embryology , Stem Cells/cytology , Stem Cells/physiology
19.
Cereb Cortex ; 27(10): 4701-4718, 2017 10 01.
Article in English | MEDLINE | ID: mdl-27620979

ABSTRACT

Loss of neurons in the neocortex is generally thought to result in a final reduction of cerebral volume. Yet, little is known on how the developing cerebral cortex copes with death of early-born neurons. Here, we tackled this issue by taking advantage of a transgenic mouse model in which, from early embryonic stages to mid-corticogenesis, abundant apoptosis is induced in the postmitotic compartment. Unexpectedly, the thickness of the mutant cortical plate at E18.5 was normal, due to an overproduction of upper layer neurons at E14.5. We developed and simulated a mathematical model to investigate theoretically the recovering capacity of the system and found that a minor increase in the probability of proliferative divisions of intermediate progenitors (IPs) is a powerful compensation lever. We confirmed experimentally that mutant mice showed an enhanced number of abventricular progenitors including basal radial glia-like cells and IPs. The latter displayed increased proliferation rate, sustained Pax6 expression and shorter cell cycle duration. Altogether, these results demonstrate the remarkable plasticity of neocortical progenitors to adapt to major embryonic insults via the modulation of abventricular divisions thereby ensuring the production of an appropriate number of neurons.


Subject(s)
Cell Proliferation/physiology , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Neurons/cytology , Animals , Cell Death , Gene Expression Regulation, Developmental/physiology , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis/physiology
20.
Cell Rep ; 17(12): 3133-3141, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28009284

ABSTRACT

Cajal-Retzius cells (CRs), the first-born neurons in the developing cerebral cortex, coordinate crucial steps in the construction of functional circuits. CRs are thought to be transient, as they disappear during early postnatal life in both mice and humans, where their abnormal persistence is associated with pathological conditions. Embryonic CRs comprise at least three molecularly and functionally distinct subtypes: septum, ventral pallium/pallial-subpallial boundary (PSB), and hem. However, whether subtype-specific features exist postnatally and through which mechanisms they disappear remain unknown. We report that CR subtypes display unique distributions and dynamics of death in the postnatal mouse cortex. Surprisingly, although all CR subtypes undergo cell death, septum, but not hem, CRs die in a Bax-dependent manner. Bax-inactivated rescued septum-CRs maintain immature electrophysiological properties. These results underlie the existence of an exquisitely refined control of developmental cell death and provide a model to test the effect of maintaining immature circuits in the adult neocortex.


Subject(s)
Cell Death/genetics , Cerebral Cortex/metabolism , Neurons/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Animals, Newborn , Cell Differentiation/genetics , Cell Lineage/genetics , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Embryo, Mammalian , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...