Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 6409, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34737295

ABSTRACT

Mutations of the mitochondrial genome (mtDNA) cause a range of profoundly debilitating clinical conditions for which treatment options are very limited. Most mtDNA diseases show heteroplasmy - tissues express both wild-type and mutant mtDNA. While the level of heteroplasmy broadly correlates with disease severity, the relationships between specific mtDNA mutations, heteroplasmy, disease phenotype and severity are poorly understood. We have carried out extensive bioenergetic, metabolomic and RNAseq studies on heteroplasmic patient-derived cells carrying the most prevalent disease related mtDNA mutation, the m.3243 A > G. These studies reveal that the mutation promotes changes in metabolites which are associated with the upregulation of the PI3K-Akt-mTORC1 axis in patient-derived cells and tissues. Remarkably, pharmacological inhibition of PI3K, Akt, or mTORC1 reduced mtDNA mutant load and partially rescued cellular bioenergetic function. The PI3K-Akt-mTORC1 axis thus represents a potential therapeutic target that may benefit people suffering from the consequences of the m.3243 A > G mutation.


Subject(s)
DNA, Mitochondrial/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , DNA, Mitochondrial/genetics , Female , Humans , Mechanistic Target of Rapamycin Complex 1/genetics , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
2.
Hum Mol Genet ; 29(19): 3224-3248, 2020 11 25.
Article in English | MEDLINE | ID: mdl-32959884

ABSTRACT

Genome-wide association studies have reported that, amongst other microglial genes, variants in TREM2 can profoundly increase the incidence of developing Alzheimer's disease (AD). We have investigated the role of TREM2 in primary microglial cultures from wild type mice by using siRNA to decrease Trem2 expression, and in parallel from knock-in mice heterozygous or homozygous for the Trem2 R47H AD risk variant. The prevailing phenotype of Trem2 R47H knock-in mice was decreased expression levels of Trem2 in microglia, which resulted in decreased density of microglia in the hippocampus. Overall, primary microglia with reduced Trem2 expression, either by siRNA or from the R47H knock-in mice, displayed a similar phenotype. Comparison of the effects of decreased Trem2 expression under conditions of lipopolysaccharide (LPS) pro-inflammatory or IL-4 anti-inflammatory stimulation revealed the importance of Trem2 in driving a number of the genes up-regulated in the anti-inflammatory phenotype. RNA-seq analysis showed that IL-4 induced the expression of a program of genes including Arg1 and Ap1b1 in microglia, which showed an attenuated response to IL-4 when Trem2 expression was decreased. Genes showing a similar expression profile to Arg1 were enriched for STAT6 transcription factor recognition elements in their promoter, and Trem2 knockdown decreased levels of STAT6. LPS-induced pro-inflammatory stimulation suppressed Trem2 expression, thus preventing TREM2's anti-inflammatory drive. Given that anti-inflammatory signaling is associated with tissue repair, understanding the signaling mechanisms downstream of Trem2 in coordinating the pro- and anti-inflammatory balance of microglia, particularly mediating effects of the IL-4-regulated anti-inflammatory pathway, has important implications for fighting neurodegenerative disease.


Subject(s)
Gene Expression Regulation , Inflammation Mediators/metabolism , Inflammation/immunology , Membrane Glycoproteins/physiology , Microglia/immunology , Mutation , Receptors, Immunologic/physiology , Transcriptome , Animals , Animals, Newborn , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/pathology , RNA-Seq , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism
3.
Nature ; 582(7811): 246-252, 2020 06.
Article in English | MEDLINE | ID: mdl-32499648

ABSTRACT

A wealth of specialized neuroendocrine command systems intercalated within the hypothalamus control the most fundamental physiological needs in vertebrates1,2. Nevertheless, we lack a developmental blueprint that integrates the molecular determinants of neuronal and glial diversity along temporal and spatial scales of hypothalamus development3. Here we combine single-cell RNA sequencing of 51,199 mouse cells of ectodermal origin, gene regulatory network (GRN) screens in conjunction with genome-wide association study-based disease phenotyping, and genetic lineage reconstruction to show that nine glial and thirty-three neuronal subtypes are generated by mid-gestation under the control of distinct GRNs. Combinatorial molecular codes that arise from neurotransmitters, neuropeptides and transcription factors are minimally required to decode the taxonomical hierarchy of hypothalamic neurons. The differentiation of γ-aminobutyric acid (GABA) and dopamine neurons, but not glutamate neurons, relies on quasi-stable intermediate states, with a pool of GABA progenitors giving rise to dopamine cells4. We found an unexpected abundance of chemotropic proliferation and guidance cues that are commonly implicated in dorsal (cortical) patterning5 in the hypothalamus. In particular, loss of SLIT-ROBO signalling impaired both the production and positioning of periventricular dopamine neurons. Overall, we identify molecular principles that shape the developmental architecture of the hypothalamus and show how neuronal heterogeneity is transformed into a multimodal neural unit to provide virtually infinite adaptive potential throughout life.


Subject(s)
Gene Expression Regulation, Developmental , Hypothalamus/cytology , Hypothalamus/embryology , Morphogenesis , Animals , Cell Differentiation , Cell Lineage , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Female , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Regulatory Networks , Genome-Wide Association Study , Glutamic Acid/metabolism , Hypothalamus/metabolism , Male , Mice , Morphogenesis/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Receptors, Immunologic/metabolism , Regulon/genetics , Signal Transduction , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism , Roundabout Proteins
4.
Brain Struct Funct ; 224(1): 277-292, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30315415

ABSTRACT

Cortical interneurons are born in the ventral forebrain and migrate tangentially in two streams at the levels of the intermediate zone (IZ) and the pre-plate/marginal zone to the developing cortex where they switch to radial migration before settling in their final positions in the cortical plate. In a previous attempt to identify the molecules that regulate stream specification, we performed transcriptomic analysis of GFP-labelled interneurons taken from the two migratory streams during corticogenesis. A number of cadherins were found to be expressed differentially, with Cadherin-8 (Cdh8) selectively present in the IZ stream. We verified this expression pattern at the mRNA and protein levels on tissue sections and found approximately half of the interneurons of the IZ expressed Cdh8. Furthermore, this cadherin was also detected in the germinal zones of the subpallium, suggesting that it might be involved not only in the migration of interneurons but also in their generation. Quantitative analysis of cortical interneurons in animals lacking the cadherin at E18.5 revealed a significant increase in their numbers. Subsequent functional in vitro experiments showed that blocking Cdh8 function led to increased cell proliferation, with the opposite results observed with over-expression, supporting its role in interneuron generation.


Subject(s)
Cadherins/metabolism , Cell Proliferation , Cerebral Cortex/metabolism , Interneurons/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Animals , Apoptosis , COS Cells , Cadherins/deficiency , Cadherins/genetics , Cerebral Cortex/embryology , Chemotaxis , Chlorocebus aethiops , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Gestational Age , Male , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Transcriptome
5.
Cereb Cortex ; 28(7): 2577-2593, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29901792

ABSTRACT

Vascular endothelial growth factor (Vegfa) is essential for promoting the vascularization of the embryonic murine forebrain. In addition, it directly influences neural development, although its role in the forming forebrain is less well elucidated. It was recently suggested that Vegfa may influence the development of GABAergic interneurons, inhibitory cells with crucial signaling roles in cortical neuronal circuits. However, the mechanism by which it affects interneuron development remains unknown. Here we investigated the developmental processes by which Vegfa may influence cortical interneuron development by analyzing transgenic mice that ubiquitously express the Vegfa120 isoform to perturb its signaling gradient. We found that interneurons reach the dorsal cortex at mid phases of corticogenesis despite an aberrant vascular network. Instead, endothelial ablation of Vegfa alters cortical interneuron numbers, their intracortical distribution and spatial proximity to blood vessels. We show for the first time that vascular-secreted guidance factors promote early-migrating interneurons in the intact forebrain in vivo and identify a novel role for vascular-Vegfa in this process.


Subject(s)
Blood Vessels/physiology , Cell Movement/genetics , GABAergic Neurons/physiology , Prosencephalon/cytology , Prosencephalon/growth & development , Vascular Endothelial Growth Factor A/metabolism , Age Factors , Animals , Blood Vessels/embryology , Chemotaxis , Computer Simulation , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Neuropilin-1/metabolism , Rats , Rats, Sprague-Dawley , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Signal Transduction/genetics , Stem Cells/physiology , Vascular Endothelial Growth Factor A/genetics
6.
Brain Struct Funct ; 222(8): 3567-3585, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28386779

ABSTRACT

Cortical interneurons are generated in the ganglionic eminences and migrate through the ventral and dorsal telencephalon before finding their final positions within the cortical plate. During early stages of migration, these cells are present in two well-defined streams within the developing cortex. In an attempt to identify candidate genes which may play a role in interneuron stream specification, we previously carried out a microarray analysis which identified a number of cadherin receptors that were differentially expressed in these streams, including Cadherin-13 (Cdh13). Expression analysis confirmed Cdh13 to be present in the preplate layer at E13.5 and, later in development, in some cortical interneurons and pyramidal cells. Analysis of Cdh13 knockout mice at E18.5, but not at E15.5, showed a reduction in the number of interneurons and late born pyramidal neurons and a concomitant increase in apoptotic cells in the cortex. These observations were confirmed in dissociated cell cultures using overexpression and short interfering RNAs (siRNAs) constructs and dominant negative inhibitory proteins. Our findings identified a novel protective role for Cdh13 in cortical neuron development.


Subject(s)
Cadherins/physiology , Cerebral Cortex/embryology , Interneurons/physiology , Animals , Apoptosis , Cadherins/genetics , Cadherins/metabolism , Cell Count , Cell Movement , Cell Proliferation , Cells, Cultured , Cerebral Cortex/metabolism , Female , Gene Expression , Interneurons/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
7.
Brain Struct Funct ; 222(5): 2217-2233, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27858201

ABSTRACT

Cortical interneurons are generated predominantly in the medial ganglionic eminence of the ventral telencephalon and migrate to the cortex during embryonic development. These cells express neuropilin (Nrp1 and Nrp2) receptors which mediate their response to the chemorepulsive class 3 semaphorin (Sema) ligands. We show here that semaphorins Sema3A and Sema3F are expressed in layers adjacent to cortical interneuron migratory streams as well as in the striatum, suggesting they may have a role in guiding these cells throughout their journey. Analysis of Sema3A -/- and Sema3F -/- mice during corticogenesis showed that absence of Sema3A, but not Sema3F, leads to aberrant migration of cortical interneurons through the striatum. Reduced number of cortical interneurons was found in the cortex of Sema3A -/-, Nrp1 -/- and Nrp2 -/- mice, as well as altered distribution in Sema3F -/-, Nrp1 -/-, Nrp2 -/- animals and especially in neuropilin double mutants. The observed decrease in interneurons in Sema3A -/- and Nrp1 -/- mice was due to altered proliferative activity of their progenitors highlighted by changes in their mitotic spindle positioning and angle of cleavage plane during cell division. These findings point to a novel role for Sema3A-Nrp1 signalling in progenitor cell dynamics and in the generation of interneurons in the ventral telencephalon.


Subject(s)
Interneurons/metabolism , Neuropilin-1/metabolism , Semaphorin-3A/metabolism , Stem Cells/metabolism , Animals , Cell Movement/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Signal Transduction/physiology
8.
J Comp Neurol ; 524(3): 518-34, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-25975775

ABSTRACT

Cortical interneurons are generated predominantly in the medial ganglionic eminence (MGE) and migrate through the ventral and dorsal telencephalon before taking their final positions within the developing cortical plate. Previously we demonstrated that interneurons from Robo1 knockout (Robo1(-/-)) mice contain reduced levels of neuropilin 1 (Nrp1) and PlexinA1 receptors, rendering them less responsive to the chemorepulsive actions of semaphorin ligands expressed in the striatum and affecting their course of migration (Hernandez-Miranda et al. [2011] J. Neurosci. 31:6174-6187). Earlier studies have highlighted the importance of Nrp1 and Nrp2 in interneuron migration, and here we assess the role of PlexinA1 in this process. We observed significantly fewer cells expressing the interneuron markers Gad67 and Lhx6 in the cortex of PlexinA1(-/-) mice compared with wild-type littermates at E14.5 and E18.5. Although the level of apoptosis was similar in the mutant and control forebrain, proliferation was significantly reduced in the former. Furthermore, progenitor cells in the MGE of PlexinA1(-/-) mice appeared to be poorly anchored to the ventricular surface and showed reduced adhesive properties, which may account for the observed reduction in proliferation. Together our data uncover a novel role for PlexinA1 in forebrain development.


Subject(s)
Cell Proliferation/physiology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/physiology , Receptors, Cell Surface/metabolism , Animals , Apoptosis/physiology , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Interneurons/cytology , Interneurons/physiology , LIM-Homeodomain Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neurogenesis/physiology , Receptors, Cell Surface/genetics , Transcription Factors/metabolism
9.
Cardiovasc Res ; 106(1): 55-66, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25691540

ABSTRACT

AIMS: The mesenchymal cushions lining the early embryonic heart undergo complex remodelling to form the membranous ventricular septum as well as the atrioventricular and semilunar valves in later life. Disruption of this process underlies the most common congenital heart defects. Here, we identified a novel role for Slit-Robo signalling in the development of the murine membranous ventricular septum and cardiac valves. METHODS AND RESULTS: Expression of Robo1 and Robo2 receptors and their ligands, Slit2 and Slit3, was present in or adjacent to all cardiac cushions/valves. Loss of Robo1 or both Robo1 and Robo2 resulted in membranous ventricular septum defects at birth, a defect also found in Slit3, but not in Slit2 mutants. Additionally, Robo1;Robo2 double mutants showed thickened immature semilunar and atrioventricular valves as well as highly penetrant bicuspid aortic valves. Slit2 mutants recapitulated the semilunar phenotype, whereas Slit3 mutants displayed thickened atrioventricular valves. Bicuspid aortic cushions were already observed at E12.5 in the Robo1;Robo2 double mutants. Expression of Notch- and downstream Hey and Hes genes was down-regulated in Robo1 mutants, suggesting that reduced Notch signalling in mice lacking Robo might underlie the defects. Luciferase assays confirmed regulation of Notch signalling by Robo. CONCLUSION: Cardiac defects in mutants for Robo or Slit range from membranous ventricular septum defects to bicuspid aortic valves. These ligands and receptors have unique functions during development of specific cardiac cushion derivatives, and the Slit-Robo signalling pathway likely enforces its role by regulating Notch signalling, making these mutants a valuable new model to study cardiac valve formation.


Subject(s)
Aortic Valve/abnormalities , Heart Defects, Congenital/genetics , Heart Valve Diseases/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Immunologic/genetics , Signal Transduction/genetics , Animals , Aortic Valve/physiopathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Bicuspid Aortic Valve Disease , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Disease Models, Animal , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Heart Defects, Congenital/physiopathology , Heart Valve Diseases/physiopathology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/physiology , Receptors, Immunologic/physiology , Receptors, Notch/genetics , Receptors, Notch/physiology , Signal Transduction/physiology , Transcription Factor HES-1 , Ventricular Septum/pathology , Roundabout Proteins
10.
Nat Commun ; 5: 4421, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25030704

ABSTRACT

Local environmental cues are indispensable for axonal growth and guidance during brain circuit formation. Here, we combine genetic and pharmacological tools, as well as systems neuroanatomy in human fetuses and mouse models, to study the role of endocannabinoid and Slit/Robo signalling in axonal growth. We show that excess 2-arachidonoylglycerol, an endocannabinoid affecting directional axonal growth, triggers corpus callosum enlargement due to the errant CB1 cannabinoid receptor-containing corticofugal axon spreading. This phenotype mechanistically relies on the premature differentiation and end-feet proliferation of CB2R-expressing oligodendrocytes. We further show the dependence of both axonal Robo1 positioning and oligodendroglial Slit2 production on cell-type-specific cannabinoid receptor activation. Accordingly, Robo1 and/or Slit2 manipulation limits endocannabinoid modulation of axon guidance. We conclude that endocannabinoids can configure focal Slit2/Robo1 signalling to modulate directional axonal growth, which may provide a basis for understanding impaired brain wiring associated with metabolic deficits and prenatal drug exposure.


Subject(s)
Brain/embryology , Brain/metabolism , Endocannabinoids/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Arachidonic Acids/pharmacology , Axons/drug effects , Axons/metabolism , Brain/drug effects , Cells, Cultured , Corpus Callosum/drug effects , Corpus Callosum/embryology , Corpus Callosum/metabolism , Female , Glycerides/pharmacology , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Pregnancy , Receptor, Cannabinoid, CB1/metabolism , Receptors, Immunologic/genetics , Roundabout Proteins
11.
J Neurosci ; 34(16): 5717-31, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24741061

ABSTRACT

The elaborate cytoarchitecture of the mammalian neocortex requires the timely production of its constituent pyramidal neurons and interneurons and their disposition in appropriate layers. Numerous chemotropic factors present in the forebrain throughout cortical development play important roles in the orchestration of these events. The Roundabout (Robo) family of receptors and their ligands, the Slit proteins, are expressed in the developing forebrain, and are known to play important roles in the generation and migration of cortical interneurons. However, few studies have investigated their function(s) in the development of pyramidal cells. Here, we observed expression of Robo1 and Slit genes (Slit1, Slit2) in cells lining the telencephalic ventricles, and found significant increases in progenitor cells (basal and apical) at embryonic day (E)12.5 and E14.5 in the developing cortex of Robo1(-/-), Slit1(-/-), and Slit1(-/-)/Slit2(-/-), but not in mice lacking the other Robo or Slit genes. Using layer-specific markers, we found that both early- and late-born pyramidal neuron populations were significantly increased in the cortices of Robo1(-/-) mice at the end of corticogenesis (E18.5). The excess number of cortical pyramidal neurons generated prenatally appears to die in early postnatal life. The observed increase in pyramidal neurons was due to prolonged proliferative activity of their progenitors and not due to changes in cell cycle events. This finding, confirmed by in utero electroporation with Robo1 short hairpin RNA (shRNA) or control constructs into progenitors along the ventricular zone as well as in dissociated cortical cell cultures, points to a novel role for Robo1 in regulating the proliferation and generation of pyramidal neurons.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Developmental/genetics , Neocortex , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/physiology , Receptors, Immunologic/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cerebral Ventricles/cytology , Cerebral Ventricles/embryology , Cerebral Ventricles/growth & development , Embryo, Mammalian , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neocortex/cytology , Neocortex/embryology , Neocortex/growth & development , Nerve Tissue Proteins/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/genetics , Roundabout Proteins
12.
Biol Open ; 2(3): 277-82, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23519094

ABSTRACT

En route to the neocortex, interneurons migrate around and avoid the developing striatum. This is due to the chemorepulsive cues of class 3 semaphorins (Sema3A and Sema3F) acting through neuropilin and plexin co-receptors expressed in interneurons. In a recent genetic screen aimed at identifying novel components that may play a role in interneuron migration, we identified LIM-kinase 2 (Limk2), a kinase previously shown to be involved in cell movement and in Sema7A-PlexinC1 signalling. Here we show that Limk2 is differentially expressed in interneurons, with a higher expression in the subpallium compared to cortex, suggesting it may play a role in their migration through the subpallium. Chemotactic assays, carried out with small interfering RNAs (siRNAs), revealed that Limk2-siRNA transfected interneurons are less responsive to Sema3A, but respond to Sema3F. Lack of responsiveness to Sema3A resulted in their aberrant invasion of the developing striatum, as demonstrated in brain slice preparations and in in utero electroporated mouse embryos with the same siRNAs. Our results reveal a previously unknown role for Limk2 in interneuron migration and Sema3A signalling.

13.
Cereb Cortex ; 23(6): 1495-508, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22661412

ABSTRACT

Laminar organization is a key feature of the mammalian cerebral cortex, but the mechanisms by which final positioning and "inside-out" distribution of neurons are determined remain largely unknown. Here, we demonstrate that Robo1, a member of the family of Roundabout receptors, regulates the correct positioning of layers II/III pyramidal neurons in the neocortex. Specifically, we used RNA interference in mice to suppress the expression of Robo1 in a subset of layers II/III neurons, and observed the positions of these cells at distinct developmental stages. In contrast to control neurons that migrated toward the pial surface by P1, Robo1-suppressed neurons exhibited a delay in entering the cortical plate at respective stages. Unexpectedly, after the first postnatal week, these neurons were predominantly located in the upper part of layers II/III, in contrast to control cells that were distributed throughout these layers. Sequential electroporation studies revealed that Robo1-suppressed cells failed to establish the characteristic inside-out neuronal distribution and, instead, they accumulated beneath the marginal zone regardless of their birthdate. These results demonstrate that Robo receptors play a crucial role in neocortical lamination and particularly in the positioning of layers II/III pyramidal neurons.


Subject(s)
Cell Movement/genetics , Cerebral Cortex/cytology , Gene Expression Regulation, Developmental/genetics , Nerve Tissue Proteins/metabolism , Pyramidal Cells/physiology , Receptors, Immunologic/metabolism , Age Factors , Animals , Animals, Newborn , COS Cells , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Chlorocebus aethiops , Deoxyuridine/analogs & derivatives , Deoxyuridine/metabolism , Female , Flow Cytometry , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Indoles/metabolism , Mice , Mice, Inbred ICR , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nuclear Proteins/metabolism , Pregnancy , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptors, Immunologic/deficiency , Repressor Proteins/metabolism , Transfection , Roundabout Proteins
14.
Circ Res ; 112(3): 465-75, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23255421

ABSTRACT

RATIONALE: The Slit-Roundabout (Robo) signaling pathway has pleiotropic functions during Drosophila heart development. However, its role in mammalian heart development is largely unknown. OBJECTIVE: To analyze the role of Slit-Robo signaling in the formation of the pericardium and the systemic venous return in the murine heart. METHODS AND RESULTS: Expression of genes encoding Robo1 and Robo2 receptors and their ligands Slit2 and Slit3 was found in or around the systemic venous return and pericardium during development. Analysis of embryos lacking Robo1 revealed partial absence of the pericardium, whereas Robo1/2 double mutants additionally showed severely reduced sinus horn myocardium, hypoplastic caval veins, and a persistent left inferior caval vein. Mice lacking Slit3 recapitulated the defects in the myocardialization, alignment, and morphology of the caval veins. Ligand binding assays confirmed Slit3 as the preferred ligand for the Robo1 receptor, whereas Slit2 showed preference for Robo2. Sinus node development was mostly unaffected in all mutants. In addition, we show absence of cross-regulation with previously identified regulators Tbx18 and Wt1. We provide evidence that pericardial defects are created by abnormal localization of the caval veins combined with ectopic pericardial cavity formation. Local increase in neural crest cell death and impaired neural crest adhesive and migratory properties underlie the ectopic pericardium formation. CONCLUSIONS: A novel Slit-Robo signaling pathway is involved in the development of the pericardium, the sinus horn myocardium, and the alignment of the caval veins. Reduced Slit3 binding in the absence of Robo1, causing impaired cardiac neural crest survival, adhesion, and migration, underlies the pericardial defects.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pericardium/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Venae Cavae/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Movement , Gene Expression Regulation, Developmental , Gestational Age , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neural Crest/abnormalities , Neural Crest/metabolism , Pericardium/abnormalities , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Sinoatrial Node/abnormalities , Sinoatrial Node/metabolism , T-Box Domain Proteins/metabolism , Tissue Culture Techniques , Venae Cavae/abnormalities , WT1 Proteins/metabolism , Roundabout Proteins
16.
Development ; 139(18): 3326-31, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22912413

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons are born in the nasal placode and migrate along olfactory and vomeronasal axons to reach the forebrain and settle in the hypothalamus, where they control reproduction. The molecular cues that guide their migration have not been fully identified, but are thought to control either cell movement directly or the patterning of their axonal substrates. Using genetically altered mouse models we show that the migration of GnRH neurons is directly modulated by Slit2 and Robo3, members of the axon guidance Slit ligand and Robo receptor families. Mice lacking Slit2 or Robo3 have a reduced number of GnRH neurons in the forebrain, but a normal complement of their supporting axons, pointing to a direct role for these molecules in GnRH neuron migration.


Subject(s)
Cell Movement/physiology , Gonadotropin-Releasing Hormone/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Animals , COS Cells , Cell Movement/genetics , Chlorocebus aethiops , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Receptors, Cell Surface
17.
Lab Invest ; 92(8): 1129-39, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22614124

ABSTRACT

Patients whose hematopoietic system is compromised by chemo- and/or radiotherapy require transplantation of hematopoietic stem and progenitor cells (HSPCs) to restore hematopoiesis. Successful homing of transplanted HSPCs to the bone marrow (BM) largely depends on their migratory potential, which is critically regulated by the chemokine CXCL12. In this study, we have investigated the expression and function of Slit proteins and their corresponding Roundabout (Robo) receptors in human HSPC migration. Slit proteins are extracellular matrix proteins that can modulate the (chemoattractant-induced) migration of mature leukocytes. We show that mRNAs for all Slits (Slit1-3) are expressed in primary BM stroma and BM-derived endothelial and stromal cell lines, but not in CD34⁺ HSPCs. Human CD34⁺ HSPCs expressed mRNAs for all Robos (Robo1-4), but only the Robo1 protein was detected on their cell surface. Functionally, Slit3 treatment increased the in vivo homing efficiency of CD34⁺ HSPCs to the BM in NOD/SCID mice, whereas Slit3-exposed HSPC migration in vitro was inhibited. These effects do not appear to result from modulated CXCL12 responsiveness as CXCR4 expression, CXCL12-induced actin polymerization or the basal and CXCL12-induced adhesion to fibronectin or BM-derived endothelial cells of CD34⁺ HSPC were not altered by Slit3 exposure. However, we show that Slit3 rapidly reduced the levels of active RhoA in HL60 cells and primary CD34⁺ HSPC, directly affecting a pathway involved in actin cytoskeleton remodeling and HSPC migration. Together, our results support a role for Slit3 in human HSPC migration in vitro and homing in vivo and might contribute to the design of future approaches aimed at improving transplantation efficiency of human CD34⁺ HSPCs.


Subject(s)
Cell Movement/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Animals , Antigens, CD/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Flow Cytometry , Humans , Membrane Proteins/genetics , Mice , Mice, SCID , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Spleen/cytology , Stem Cell Transplantation , rhoA GTP-Binding Protein/metabolism , Roundabout Proteins
18.
Eur J Neurosci ; 34(10): 1584-94, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22103416

ABSTRACT

Cortical interneurons originate in the ganglionic eminences of the subpallium and migrate into the cortex in well-defined tangential streams. At the start of corticogenesis, two streams of migrating neurons are evident: a superficial one at the level of the preplate (PPL), and a deeper one at the level of the intermediate zone (IZ). Currently, little is known about the signalling mechanisms that regulate interneuron migration, and almost nothing is known about the molecules that may be involved in their choice of migratory stream. Here, we performed a microarray analysis, comparing the changes in gene expression between cells migrating in the PPL and those migrating in the IZ at embryonic day 13.5. This analysis identified genes, many of them novel, that were upregulated in one of the two streams. Moreover, polymerase chain reaction, in situ hybridization experiments and immunohistochemistry showed the expression of these genes in interneurons migrating within the PPL or IZ, suggesting that they play a role in their migration and choice of stream.


Subject(s)
Cell Movement/physiology , Cerebral Cortex , Gene Expression , Interneurons/physiology , Signal Transduction/physiology , Animals , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Gene Expression Profiling , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Interneurons/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
J Neurosci ; 31(16): 6174-87, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21508241

ABSTRACT

Cortical interneurons, generated predominantly in the medial ganglionic eminence, migrate around and avoid the developing striatum in the subpallium en route to the cortex. This is attributable to the chemorepulsive cues of class 3 semaphorins expressed in the striatal mantle and acting through neuropilin (Nrp1 and Nrp2) receptors expressed in these cells. Cortical interneurons also express Robo receptors, and we show here that in mice lacking Robo1, but not Robo2, these cells migrate aberrantly through the striatum. In vitro experiments demonstrated that interneurons lacking Robo1 function are significantly less responsive to the effects of semaphorins. Failure to respond to semaphorin appears to be attributable to a reduction in Nrp1 and PlexinA1 receptors within these cells. Biochemical studies further demonstrated that Robo1 binds directly to Nrp1, but not to semaphorins, and this interaction is mediated by a region contained within its first two Ig domains. Thus, we show for the first time that Robo1 interacts with Nrp1 to modulate semaphorin signaling in the developing forebrain and direct the migration of interneurons through the subpallium and into the cortex.


Subject(s)
Cerebral Cortex/metabolism , Chemotaxis/physiology , Interneurons/metabolism , Nerve Tissue Proteins/metabolism , Prosencephalon/metabolism , Receptors, Immunologic/metabolism , Semaphorins/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Cell Line , Cells, Cultured , Cerebral Cortex/cytology , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Interneurons/cytology , Mice , Mice, Knockout , Neuropilin-1/metabolism , Neuropilin-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Roundabout Proteins
20.
Development ; 136(18): 3173-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19700621

ABSTRACT

Elucidating the gene regulatory networks that govern pharyngeal arch artery (PAA) development is an important goal, as such knowledge can help to identify new genes involved in cardiovascular disease. The transcription factor Tbx1 plays a vital role in PAA development and is a major contributor to cardiovascular disease associated with DiGeorge syndrome. In this report, we used various genetic approaches to reveal part of a signalling network by which Tbx1 controls PAA development in mice. We investigated the crucial role played by the homeobox-containing transcription factor Gbx2 downstream of Tbx1. We found that PAA formation requires the pharyngeal surface ectoderm as a key signalling centre from which Gbx2, in response to Tbx1, triggers essential directional cues to the adjacent cardiac neural crest cells (cNCCs) en route to the caudal PAAs. Abrogation of this signal generates cNCC patterning defects leading to PAA abnormalities. Finally, we showed that the Slit/Robo signalling pathway is activated during cNCC migration and that components of this pathway are affected in Gbx2 and Tbx1 mutant embryos at the time of PAA development. We propose that the spatiotemporal control of this tightly orchestrated network of genes participates in crucial aspects of PAA development.


Subject(s)
Arteries/embryology , Body Patterning/physiology , Branchial Region , Cell Movement/physiology , Ectoderm , Homeodomain Proteins/metabolism , Neural Crest/cytology , T-Box Domain Proteins/metabolism , Animals , Arteries/abnormalities , Arteries/anatomy & histology , Branchial Region/abnormalities , Branchial Region/blood supply , Branchial Region/embryology , Ectoderm/anatomy & histology , Ectoderm/embryology , Ectoderm/metabolism , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Glycoproteins/metabolism , Heart/embryology , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/physiology , T-Box Domain Proteins/genetics , Roundabout Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...