Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cell Rep ; 40(5): 111157, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35926459

ABSTRACT

The function of the cerebral cortex depends on various types of interneurons (cortical interneurons [cINs]) and their appropriate allocation to the cortical layers. Caudal ganglionic eminence-derived cINs (cGE-cINs) are enriched in superficial layers. Developmental mechanisms directing cGE-cINs toward superficial layers remain poorly understood. We examine how developmental and final positioning of cGE-cINs are influenced by the Cxcl12, Cxcr4, Ackr3 module, the chief attractant system guiding medial ganglionic eminence-derived cINs (mGE-cINs). We find that Cxcl12 attracts cGE-cINs through Cxcr4 and supports their layer-specific positioning in the developing cortex. This requires the prevention of excessive Cxcr4 stimulation by Ackr3-mediated Cxcl12 sequestration. Postnatally, Ackr3 confines Cxcl12 action to the marginal zone. Unlike mGE-cINs, cGE-cINs continue to express Cxcr4 at early postnatal stages, which permits cGE-cINs to become positioned in the forming layer 1. Thus, chemoattraction by Cxcl12 guides cGE-cINs and holds them in superficial cortical layers.


Subject(s)
Cerebral Cortex , Interneurons , Cell Movement/physiology , Cerebral Cortex/physiology , Interneurons/physiology , Median Eminence , Mesoderm
2.
PLoS Genet ; 17(3): e1009441, 2021 03.
Article in English | MEDLINE | ID: mdl-33739968

ABSTRACT

Biallelic mutations in DONSON, an essential gene encoding for a replication fork protection factor, were linked to skeletal abnormalities and microcephaly. To better understand DONSON function in corticogenesis, we characterized Donson expression and consequences of conditional Donson deletion in the mouse telencephalon. Donson was widely expressed in the proliferation and differentiation zones of the embryonic dorsal and ventral telencephalon, which was followed by a postnatal expression decrease. Emx1-Cre-mediated Donson deletion in progenitors of cortical glutamatergic neurons caused extensive apoptosis in the early dorsomedial neuroepithelium, thus preventing formation of the neocortex and hippocampus. At the place of the missing lateral neocortex, these mutants exhibited a dorsal extension of an early-generated paleocortex. Targeting cortical neurons at the intermediate progenitor stage using Tbr2-Cre evoked no apparent malformations, whereas Nkx2.1-Cre-mediated Donson deletion in subpallial progenitors ablated 75% of Nkx2.1-derived cortical GABAergic neurons. Thus, the early telencephalic neuroepithelium depends critically on Donson function. Our findings help explain why the neocortex is most severely affected in individuals with DONSON mutations and suggest that DONSON-dependent microcephaly might be associated with so far unrecognized defects in cortical GABAergic neurons. Targeting Donson using an appropriate recombinase is proposed as a feasible strategy to ablate proliferating and nascent cells in experimental research.


Subject(s)
Cell Cycle Proteins/genetics , Cell Differentiation , Cerebral Cortex/metabolism , GABAergic Neurons/metabolism , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism , Neurons/metabolism , Nuclear Proteins/genetics , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cerebral Cortex/cytology , GABAergic Neurons/cytology , Hippocampus/metabolism , Mice , Neocortex , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/cytology , Telencephalon/cytology , Telencephalon/metabolism
3.
Cell Rep ; 26(6): 1473-1488.e9, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30726732

ABSTRACT

Phosphorylation of heptahelical receptors is thought to regulate G protein signaling, receptor endocytosis, and non-canonical signaling via recruitment of ß-arrestins. We investigated chemokine receptor functionality under phosphorylation-deficient and ß-arrestin-deficient conditions by studying interneuron migration in the embryonic cortex. This process depends on CXCL12, CXCR4, G protein signaling and on the atypical CXCL12 receptor ACKR3. We found that phosphorylation was crucial, whereas ß-arrestins were dispensable for ACKR3-mediated control of CXCL12 levels in vivo. Cortices of mice expressing phosphorylation-deficient ACKR3 exhibited a major interneuron migration defect, which was accompanied by excessive activation and loss of CXCR4. Cxcl12-overexpressing mice mimicked this phenotype. Excess CXCL12 caused lysosomal CXCR4 degradation, loss of CXCR4 responsiveness, and, ultimately, similar motility defects as Cxcl12 deficiency. By contrast, ß-arrestin deficiency caused only a subtle migration defect mimicked by CXCR4 gain of function. These findings demonstrate that phosphorylation regulates atypical chemokine receptor function without ß-arrestin involvement in chemokine sequestration and non-canonical signaling.


Subject(s)
Cell Movement , Interneurons/metabolism , Receptors, CXCR/metabolism , Animals , CHO Cells , Chemokine CXCL12/metabolism , Cricetinae , Cricetulus , HEK293 Cells , Humans , Interneurons/physiology , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Receptors, CXCR/genetics , beta-Arrestins/metabolism
4.
Curr Biol ; 29(2): 332-339.e5, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30639110

ABSTRACT

Neurons of the neocortex are organized into six radial layers, which have appeared at different times during evolution, with the superficial layers representing a more recent acquisition. Input to the neocortex predominantly reaches superficial layers (SL, i.e., layers (L) 2-4), while output is generated in deep layers (DL, i.e., L5-6) [1]. Intracortical connections, which bridge input and output pathways, are key components of cortical circuits because they allow the propagation and processing of information within the neocortex. Two main types of intracortically projecting neurons (ICPN) can be distinguished by their axonal features: L4 spiny stellate neurons (SSN) with short axons projecting locally within cortical columns [2-5], and SL and DL long-range projection neurons, including callosally projecting neurons (CPNSL and CPNDL) [5, 6]. Here, we investigate the molecular hallmarks that distinguish SSN, CPNSL, and CPNDL and relate their transcriptional signatures with their output connectivity. Specifically, taking advantage of the presence of CPN in both SL and DL, we identify lamina-independent genetic hallmarks of a constant projection motif (i.e., interhemispheric projection). By performing unbiased transcriptomic comparisons between CPNSL, CPNDL and SSN, we provide specific molecular profiles for each of these populations and show that target identity supersedes laminar position in defining ICPN transcriptional diversity. Together, these findings reveal a projection-based organization of transcriptional programs across cortical layers, which we propose reflects conserved strategy to protect canonical circuit structure (and hence function) across a diverse range of neuroanatomies.


Subject(s)
Neocortex/physiology , Neural Pathways/physiology , Neurons/physiology , Animals , Axons/physiology , Female , Male , Mice, Inbred C57BL , Neurons/classification , Rats
5.
Glia ; 66(8): 1566-1576, 2018 08.
Article in English | MEDLINE | ID: mdl-29537098

ABSTRACT

Adult hippocampal neurogenesis is implicated in learning and memory processing. It is tightly controlled at several levels including progenitor proliferation as well as migration, differentiation and integration of new neurons. Hippocampal progenitors and immature neurons reside in the subgranular zone (SGZ) and are equipped with the CXCL12-receptor CXCR4 which contributes to defining the SGZ as neurogenic niche. The atypical CXCL12-receptor CXCR7 functions primarily by sequestering extracellular CXCL12 but whether CXCR7 is involved in adult neurogenesis has not been assessed. We report that granule neurons (GN) upregulate CXCL12 and CXCR7 during dentate gyrus maturation in the second postnatal week. To test whether GN-derived CXCL12 regulates neurogenesis and if neuronal CXCR7 receptors influence this process, we conditionally deleted Cxcl12 and Cxcr7 from the granule cell layer. Cxcl12 deletion resulted in lower numbers, increased dispersion and abnormal dendritic growth of immature GN and reduced neurogenesis. Cxcr7 ablation caused an increase in progenitor proliferation and progenitor numbers and reduced dispersion of immature GN. Thus, we provide a new mechanism where CXCL12-signals from GN prevent dispersion and support maturation of newborn GN. CXCR7 receptors of GN modulate the CXCL12-mediated feedback from GN to the neurogenic niche.


Subject(s)
Chemokine CXCL12/metabolism , Dentate Gyrus/metabolism , Neurogenesis/physiology , Neurons/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Dendrites/metabolism , Hippocampus/metabolism , Mice, Transgenic , Neural Stem Cells/metabolism
6.
J Neurosci ; 35(38): 13053-63, 2015 Sep 23.
Article in English | MEDLINE | ID: mdl-26400936

ABSTRACT

Glutamatergic principal neurons, GABAergic interneurons and thalamocortical axons (TCAs) are essential elements of the cerebrocortical network. Principal neurons originate locally from radial glia and intermediate progenitors (IPCs), whereas interneurons and TCAs are of extrinsic origin. Little is known how the assembly of these elements is coordinated. C-X-C motif chemokine 12 (CXCL12), which is known to guide axons outside the neural tube and interneurons in the cortex, is expressed in the meninges and IPCs. Using mouse genetics, we dissected the influence of IPC-derived CXCL12 on TCAs and interneurons by showing that Cxcl12 ablation in IPCs, leaving meningeal Cxcl12 intact, attenuates intracortical TCA growth and disrupts tangential interneuron migration in the subventricular zone. In accordance with strong CXCR4 expression in the forming thalamus and TCAs, we identified a CXCR4-dependent growth-promoting effect of CXCL12 on TCAs in thalamus explants. Together, our findings indicate a cell-autonomous role of CXCR4 in promoting TCA growth. We propose that CXCL12 signals from IPCs link cortical neurogenesis to the progression of TCAs and interneurons spatially and temporally. Significance statement: The cerebral cortex exerts higher brain functions including perceptual and emotional processing. Evolutionary expansion of the mammalian cortex is mediated by intermediate progenitors, transient amplifying cells generating cortical excitatory neurons. During the peak period of cortical neurogenesis, migrating precursors of inhibitory interneurons originating in subcortical areas and thalamic axons invade the cortex. Although defects in the assembly of cortical network elements cause neurological and mental disorders, little is known how neurogenesis, interneuron recruitment, and axonal ingrowth are coordinated. We demonstrate that intermediate progenitors release the chemotactic cytokine CXCL12 to promote intracortical interneuron migration and growth of thalamic axons via the cognate receptor CXCR4. This paracrine signal may ensure thalamocortical connectivity and dispersion of inhibitory neurons in the rapidly growing cortex.


Subject(s)
Cerebral Cortex/cytology , Chemokine CXCL12/metabolism , Interneurons/physiology , Signal Transduction/physiology , Stem Cells/physiology , Thalamus/cytology , Animals , Axons/metabolism , Cerebral Cortex/embryology , Chemokine CXCL12/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Intermediate Filaments/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins , Neural Pathways/physiology , Organ Culture Techniques , Receptors, CXCR/genetics , Receptors, CXCR/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Thalamus/embryology
7.
Development ; 141(9): 1857-63, 2014 May.
Article in English | MEDLINE | ID: mdl-24718993

ABSTRACT

The CXCL12/CXCR4 signaling pathway is involved in the development of numerous neuronal and non-neuronal structures. Recent work established that the atypical second CXCL12 receptor, CXCR7, is essential for the proper migration of interneuron precursors in the developing cerebral cortex. Two CXCR7-mediated functions were proposed in this process: direct modulation of ß-arrestin-mediated signaling cascades and CXCL12 scavenging to regulate local chemokine availability and ensure responsiveness of the CXCL12/CXCR4 pathway in interneurons. Neither of these functions has been proven in the embryonic brain. Here, we demonstrate that migrating interneurons efficiently sequester CXCL12 through CXCR7. CXCR7 ablation causes excessive phosphorylation and downregulation of CXCR4 throughout the cortex in mice expressing CXCL12, but not in CXCL12-deficient animals. Cxcl12(-/-) mice lack activated CXCR4 in embryonic brain lysates and display a similar interneuron positioning defect as Cxcr4(-/-), Cxcr7(-/-) and Cxcl12(-/-);Cxcr7(-/-) animals. Thus, CXCL12 is the only CXCR4-activating ligand in the embryonic brain and deletion of one of the CXCL12 receptors is sufficient to generate a migration phenotype that corresponds to the CXCL12-deficient pathway. Our findings imply that interfering with the CXCL12-scavenging activity of CXCR7 causes loss of CXCR4 function as a consequence of excessive CXCL12-mediated CXCR4 activation and degradation.


Subject(s)
Cell Movement , Cerebral Cortex/cytology , Chemokine CXCL12/metabolism , Down-Regulation , Interneurons/cytology , Interneurons/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Animals , Embryo, Mammalian/cytology , HEK293 Cells , Humans , Mice , Models, Biological , Recombinant Fusion Proteins/metabolism
8.
Development ; 141(3): 661-73, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24449842

ABSTRACT

The proper functioning of the dopaminergic system requires the coordinated formation of projections extending from dopaminergic neurons in the substantia nigra (SN), ventral tegmental area (VTA) and retrorubral field to a wide array of forebrain targets including the striatum, nucleus accumbens and prefrontal cortex. The mechanisms controlling the assembly of these distinct dopaminergic cell clusters are not well understood. Here, we have investigated in detail the migratory behavior of dopaminergic neurons giving rise to either the SN or the medial VTA using genetic inducible fate mapping, ultramicroscopy, time-lapse imaging, slice culture and analysis of mouse mutants. We demonstrate that neurons destined for the SN migrate first radially and then tangentially, whereas neurons destined for the medial VTA undergo primarily radial migration. We show that tangentially migrating dopaminergic neurons express the components of the reelin signaling pathway, whereas dopaminergic neurons in their initial, radial migration phase express CXC chemokine receptor 4 (CXCR4), the receptor for the chemokine CXC motif ligand 12 (CXCL12). Perturbation of reelin signaling interferes with the speed and orientation of tangentially, but not radially, migrating dopaminergic neurons and results in severe defects in the formation of the SN. By contrast, CXCR4/CXCL12 signaling modulates the initial migration of dopaminergic neurons. With this study, we provide the first molecular and functional characterization of the distinct migratory pathways taken by dopaminergic neurons destined for SN and VTA, and uncover mechanisms that regulate different migratory behaviors of dopaminergic neurons.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement , Chemokine CXCL12/metabolism , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Extracellular Matrix Proteins/metabolism , Nerve Tissue Proteins/metabolism , Serine Endopeptidases/metabolism , Animals , Cell Lineage , Embryonic Development , Ligands , Mice , Mice, Knockout , Models, Biological , Receptors, CXCR4/metabolism , Reelin Protein , Signal Transduction , Substantia Nigra/cytology , Time-Lapse Imaging , Ventral Tegmental Area/cytology
9.
J Neurosci ; 33(44): 17527-37, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24174685

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons are neuroendocrine cells, located in the hypothalamus, that play an essential role in mammalian reproduction. These neurons originate in the nasal placode and migrate during embryonic development, in association with olfactory/vomeronasal nerves, first in the nose, then through the cribriform plate to enter the forebrain, before settling in the hypothalamus. One of the molecules required for their early migration in the nose is the chemokine CXCL12, which is expressed in the embryonic nasal mesenchyme in an increasing ventral to dorsal gradient, presumably guiding GnRH neurons toward the forebrain. Mice lacking CXCR4, the receptor for CXCL12, exhibit defective GnRH cell movement and a significant reduction in their number, suggesting that CXCL12/CXCR4 signaling is important in the migration and survival of these neurons. Here, we investigated the role of the more recently identified second CXCL12 receptor, CXCR7, in GnRH neuron development. We demonstrate that CXCR7 is expressed along the migratory path of GnRH neurons in the nasal cavity and, although not expressed by GnRH neurons, it affects their migration as indicated by the ectopic accumulation of these cells in the nasal compartment in CXCR7(-/-) mice. Absence of CXCR7 caused abnormal accumulation of CXCL12-RFP at CXCR4-positive sites in the nasal area of CXCL12-RFP-transgenic mice and excessive CXCL12-dependent intracellular clustering of CXCR4 in GnRH neurons, suggesting internalization. These findings imply that CXCR7 regulates CXCL12 availability by acting as a scavenger along the migratory path of GnRH neurons and, thus, influences the migration of these cells in a noncell-autonomous manner.


Subject(s)
Cell Movement/physiology , Chemokine CXCL12/genetics , Gonadotropin-Releasing Hormone/physiology , Neurons/cytology , Neurons/physiology , Receptors, CXCR/genetics , Receptors, CXCR/physiology , Animals , Chemokine CXCL12/biosynthesis , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Receptors, CXCR/deficiency , Receptors, CXCR4/deficiency , Receptors, CXCR4/genetics
10.
Hippocampus ; 23(12): 1345-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23929505

ABSTRACT

Neurogenesis in the adult dentate gyrus (DG) generates new granule neurons that differentiate in the inner one-third of the granule cell layer (GCL). The migrating precursors of these neurons arise from neural stem cells (NSCs) in the subgranular zone (SGZ). Although it is established that pathological conditions, including epilepsy and stroke, cause dispersion of granule neuron precursors, little is known about the factors that regulate their normal placement. Based on the high expression of the chemokine CXCL12 in the adult GCL and its role in guiding neuronal migration in development, we addressed the function of the CXCL12 receptor CXCR4 in adult neurogenesis. Using transgenic reporter mice, we detected Cxcr4-GFP expression in NSCs, neuronal-committed progenitors, and immature neurons of adult and aged mice. Analyses of hippocampal NSC cultures and hippocampal tissue by immunoblot and immunohistochemistry provided evidence for CXCL12-promoted phosphorylation/activation of CXCR4 receptors in NSCs in vivo and in vitro. Cxcr4 deletion in NSCs of the postnatal or mature DG using Cre technology reduced neurogenesis. Fifty days after Cxcr4 ablation in the mature DG, the SGZ showed a severe reduction of Sox2-positive neural stem/early progenitor cells, NeuroD-positive neuronal-committed progenitors, and DCX-positive immature neurons. Many immature neurons were ectopically placed in the hilus and inner molecular layer, and some developed an aberrant dendritic morphology. Only few misplaced cells survived permanently as ectopic neurons. Thus, CXCR4 signaling maintains the NSC pool in the DG and specifies the inner one-third of the GCL as differentiation area for immature granule neurons.


Subject(s)
Dentate Gyrus/cytology , Gene Expression Regulation, Developmental/physiology , Neurons/physiology , Receptors, CXCR4/metabolism , Age Factors , Animals , Anti-HIV Agents/pharmacology , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Benzylamines , Cell Differentiation/drug effects , Cells, Cultured , Chemokine CXCL12/pharmacology , Cyclams , Doublecortin Domain Proteins , Doublecortin Protein , Gene Expression Regulation, Developmental/genetics , Heterocyclic Compounds/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neuropeptides/metabolism , Receptors, CXCR4/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...