Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Type of study
Publication year range
1.
Neuroscience ; 519: 162-176, 2023 05 21.
Article in English | MEDLINE | ID: mdl-36990270

ABSTRACT

Abnormal N-methyl-D-aspartate receptor (NMDAr) function has been linked to oscillopathies, psychosis, and cognitive dysfunction in schizophrenia (SCZ). Here, we investigate the role of NMDAr hypofunction in pathological oscillations and behavior. We implanted mice with tetrodes in the dorsal/intermediate hippocampus and medial prefrontal cortex (mPFC), administered the NMDAr antagonist MK-801, and recorded oscillations during spontaneous exploration in an open field and in the y-maze spatial working memory test. Our results show that NMDAr blockade disrupted the correlation between oscillations and speed of movement, crucial for internal representations of distance. In the hippocampus, MK-801 increased gamma oscillations and disrupted theta/gamma coupling during spatial working memory. In the mPFC, MK-801 increased the power of theta and gamma, generated high-frequency oscillations (HFO 155-185 Hz), and disrupted theta/gamma coupling. Moreover, the performance of mice in the spatial working memory version of the y-maze was strongly correlated with CA1-PFC theta/gamma co-modulation. Thus, theta/gamma mediated by NMDAr function might explain several of SCZ's cognitive symptoms and might be crucial to explaining hippocampal-PFC interaction.


Subject(s)
Dizocilpine Maleate , Memory, Short-Term , Mice , Animals , Dizocilpine Maleate/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Prefrontal Cortex/metabolism , Theta Rhythm/physiology , Hippocampus/metabolism , Memory Disorders/chemically induced , Spatial Memory
2.
Neuroimage ; 97: 95-106, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24769183

ABSTRACT

Genetic mouse models of neurodevelopmental disorders are being massively generated, but technologies for their high-throughput phenotyping are missing. The potential of high-resolution magnetic resonance imaging (MRI) for structural phenotyping has been demonstrated before. However, application to the embryonic mouse central nervous system has been limited by the insufficient anatomical detail. Here we present a method that combines staining of live embryos with a contrast agent together with MR microscopy after fixation, to provide unprecedented anatomical detail at relevant embryonic stages. By using this method we have phenotyped the embryonic forebrain of Robo1/2(-/-) double mutant mice enabling us to identify most of the well-known anatomical defects in these mutants, as well as novel more subtle alterations. We thus demonstrate the potential of this methodology for a fast and reliable screening of subtle structural abnormalities in the developing mouse brain, as those associated to defects in disease-susceptibility genes of neurologic and psychiatric relevance.


Subject(s)
Central Nervous System/embryology , Embryo, Mammalian/anatomy & histology , Magnetic Resonance Imaging/methods , Animals , Brain/embryology , Contrast Media , Embryonic Development/physiology , Female , Heterocyclic Compounds , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Microscopy , Organometallic Compounds , Phenotype , Pregnancy
3.
Neurobiol Aging ; 29(4): 542-53, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17169463

ABSTRACT

The function of the amyloid precursor protein (APP), a key molecule in Alzheimer's disease (AD) remains unknown. Among the proteins that interact with the APP cytoplasmic domain in vitro and in heterologous systems is Disabled-1, a signaling molecule of the reelin pathway. The physiological consequence of this interaction is unknown. Here we used an in vitro model of hippocampal neurons grown on a reelin substrate that inhibits neurite outgrowth. Our results show that an excess of APP cytoplasmic domain internalized by a cell permeable peptide, is able to antagonize the neurite outgrowth inhibition of reelin. The APP cytoplasmic domain binds Disabled-1 and retains it in the cytoplasm, preventing it from reaching the plasma membrane and sequesters tyrosine phosphorylated Disabled-1, both of which disrupt reelin signaling. In the context of AD, increased formation of APP cytoplasmic domain in the cytosol released after cleavage of the A beta peptide, could then inhibit reelin signaling pathway in the hippocampus and thus influence synaptic plasticity.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/physiology , Cytoplasm/physiology , Extracellular Matrix Proteins/antagonists & inhibitors , Extracellular Matrix Proteins/physiology , Hippocampus/growth & development , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Neurites/physiology , Serine Endopeptidases/physiology , Amino Acid Sequence , Amyloid beta-Protein Precursor/chemistry , Animals , COS Cells , Cell Membrane Permeability/physiology , Cells, Cultured , Chlorocebus aethiops , Cytoplasm/chemistry , Mice , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Neural Inhibition/physiology , Neurons/physiology , Protein Structure, Tertiary/physiology , Rats , Reelin Protein
4.
Development ; 127(22): 4797-810, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11044395

ABSTRACT

Diffusible factors, including netrins and semaphorins, are believed to be important cues for the formation of neural circuits in the forebrain. Here we have examined the role of netrin 1 in the development of hippocampal connections. We show that netrin 1 and its receptor, Dcc, are expressed in the developing fimbria and in projection neurons, respectively, and that netrin 1 promotes the outgrowth of hippocampal axons in vitro via DCC receptors. We also show that the hippocampus of netrin 1-deficient mice shows a misorientation of fiber tracts and pathfinding errors, as detected with antibodies against the surface proteins TAG-1, L1 and DCC. DiI injections show that hippocampal commissural axons do not cross the midline in these mutants. Instead, when axons approach the midline, they turn ventrally and form a massive aberrant projection to the ipsilateral septum. In addition, both the ipsilateral entorhino-hippocampal and the CA3-to-CA1 associational projections show an altered pattern of layer-specific termination in netrin 1-deficient mice. Finally, optical recordings with the Ca(2+) indicator Fura 2-AM show that spontaneous neuronal activity is reduced in the septum of netrin 1-mutant mice. We conclude that netrin 1 is required not only for the formation of crossed connections in the forebrain, but also for the appropriate layer-specific targeting of ipsilateral projections and for the control of normal levels of spontaneous neural activity.


Subject(s)
Hippocampus/abnormalities , Nerve Growth Factors/deficiency , Tumor Suppressor Proteins , Animals , Animals, Newborn , Axons/ultrastructure , Calcium Signaling , Cell Adhesion Molecules/physiology , DCC Receptor , Gene Expression Regulation, Developmental , Hippocampus/growth & development , Hippocampus/physiopathology , In Vitro Techniques , Mice , Mice, Knockout , Mice, Mutant Strains , Nerve Growth Factors/genetics , Nerve Growth Factors/physiology , Netrin-1 , Neural Pathways/abnormalities , Neural Pathways/growth & development , Neural Pathways/physiopathology , Receptors, Cell Surface
5.
Development ; 126(16): 3637-48, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10409509

ABSTRACT

Extracellular matrix (ECM) proteins play an important role in early cortical development, specifically in the formation of neural connections and in controlling the cyto-architecture of the central nervous system. F-spondin and Mindin are a family of matrix-attached adhesion molecules that share structural similarities and overlapping domains of expression. Genes for both proteins contain a thrombospondin type I repeat(s) at the C terminus and an FS1-FS2 (spondin) domain. Both the vertebrate F-spondin and the zebrafish mindins are expressed on the embryonic floor plate. In the current study we have cloned the rat homologue of mindin and studied its expression and activity together with F-spondin in the developing rodent brain. The two genes are abundantly expressed in the developing hippocampus. In vitro studies indicate that both F-spondin and Mindin promote adhesion and outgrowth of hippocampal embryonic neurons. We have also demonstrated that the two proteins bind to a putative receptor(s) expressed on both hippocampal and sensory neurons.


Subject(s)
Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Developmental , Growth Substances , Hippocampus/embryology , Neural Cell Adhesion Molecules/genetics , Neurons/physiology , Peptides , Zebrafish Proteins , Amino Acid Sequence , Animals , Cloning, Molecular , Drosophila , Drosophila Proteins , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/physiology , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Molecular Sequence Data , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/physiology , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Repetitive Sequences, Amino Acid , Sequence Alignment , Sequence Homology, Amino Acid , Transcription, Genetic , Zebrafish
6.
Exp Neurol ; 156(2): 268-82, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10328935

ABSTRACT

Reelin is a large, extracellular matrix protein involved in neuronal migration and axonal growth. To analyze the contribution of Reelin to the development of the commissural projection in the hippocampus, we analyzed the ontogeny of this projection in the reeler mutant mouse. Injections of the lipophilic tracer DiI revealed many commissural fibers in the hippocampus of both reeler and control mice at P1-P2. At P5, at P12, and in the adult, the topography of commissural connections was normal in the CA1 region of reeler mice, with axons innervating the stratum radiatum and stratum oriens. In contrast, in the CA3/CA2 region, commissural fibers abnormally innervated the stratum lacunosum-moleculare and, in the dentate gyrus, some fibers were observed in the outer molecular layer. Next, we monitored the distribution of Cajal-Retzius cells in the hippocampus of reeler mutant mice and noted that the stratum lacunosum-moleculare of the CA3/CA2 region was largely devoid of Cajal-Retzius (CR) cells. Taken together, the above results indicate that in the absence of CR cells in the CA3/CA2, commissural axons abnormally grow to the stratum lacunosum-moleculare. To test this hypothesis a series of coculture experiments was performed in collagen gels, in which the CA3 axonal growth was monitored when confronted to the marginal zone. These experiments showed that the marginal zone containing CR cells exerts short-range inhibitory influences for commissural axonal growth.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Hippocampus/pathology , Mice, Neurologic Mutants/anatomy & histology , Nerve Tissue Proteins/physiology , Age Factors , Animals , Axons/ultrastructure , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Coculture Techniques , Crosses, Genetic , Dentate Gyrus/chemistry , Dentate Gyrus/pathology , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Genotype , Hippocampus/chemistry , In Situ Hybridization , Male , Mice , Mice, Inbred BALB C , Mice, Neurologic Mutants/genetics , Morphogenesis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Reelin Protein , Serine Endopeptidases
7.
J Neurosci ; 19(4): 1345-58, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-9952412

ABSTRACT

Here we examine the role of Reelin, an extracellular protein involved in neuronal migration, in the formation of hippocampal connections. Both at prenatal and postnatal stages, the general laminar and topographic distribution of entorhinal projections is preserved in the hippocampus of reeler mutant mice, in the absence of Reelin. However, developing and adult entorhinal afferents show severe alterations, including increased numbers of misrouted fibers and the formation of abnormal patches of termination from the medial and lateral entorhinal cortices. At perinatal stages, single entorhinal axons in reeler mice are grouped into thick bundles, and they have decreased axonal branching and decreased extension of axon collaterals. We also show that the number of entorhino-hippocampal synapses is lower in reeler mice than in control animals during development. Studies performed in mixed entorhino-hippocampal co-cultures combining slices from reeler and wild-type mice indicate that these abnormalities are caused by the lack of Reelin in the target hippocampus. These findings imply that Reelin fulfills a modulatory role during the formation of layer-specific and topographic connections in the hippocampus. They also suggest that Reelin promotes maturation of single fibers and synaptogenesis by entorhinal afferents.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Entorhinal Cortex/growth & development , Extracellular Matrix Proteins/physiology , Hippocampus/growth & development , Nerve Tissue Proteins/metabolism , Synapses/physiology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Entorhinal Cortex/cytology , Entorhinal Cortex/ultrastructure , Extracellular Matrix Proteins/genetics , Hippocampus/cytology , Hippocampus/ultrastructure , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Neurologic Mutants , Microscopy, Electron , Nerve Fibers/metabolism , Neural Pathways/cytology , Neural Pathways/growth & development , Neural Pathways/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reelin Protein , Serine Endopeptidases , Synapses/ultrastructure
8.
Development ; 125(21): 4313-23, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9753685

ABSTRACT

The semaphorins are the largest family of repulsive axon guidance molecules. Secreted semaphorins bind neuropilin receptors and repel sensory, sympathetic and motor axons. Here we show that CA1, CA3 and dentate gyrus axons from E15-E17 mouse embryo explants are selectively repelled by entorhinal cortex and neocortex. The secreted semaphorins Sema III and Sema IV and their receptors Neuropilin-1 and -2 are expressed in the hippocampal formation during appropriate stages. Sema III and Sema IV strongly repel CA1, CA3 and dentate gyrus axons; entorhinal axons are only repelled by Sema III. An antibody against Neuropilin-1 blocks the repulsive action of Sema III and the entorhinal cortex, but has no effect on Sema IV-induced repulsion. Thus, chemorepulsion plays a role in axon guidance in the hippocampus, secreted semaphorins are likely to be responsible for this action, and the same axons can be repelled by two distinct semaphorins via two different receptors.


Subject(s)
Axons/physiology , Chemotaxis , Glycoproteins/physiology , Hippocampus/physiology , Nerve Growth Factors/physiology , Animals , COS Cells , Chemotactic Factors/genetics , Chemotactic Factors/metabolism , Chemotactic Factors/physiology , Coculture Techniques , Entorhinal Cortex/metabolism , Entorhinal Cortex/physiology , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Glycoproteins/metabolism , Hippocampus/embryology , Hippocampus/metabolism , In Situ Hybridization , Mice , Neocortex/physiology , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Netrin-1 , Neuropilin-1 , RNA, Messenger/metabolism , Semaphorin-3A , Transfection , Tumor Suppressor Proteins
9.
J Neurosci ; 18(18): 7336-50, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9736654

ABSTRACT

Recent studies have suggested a role for neurotrophins in the growth and refinement of neural connections, in dendritic growth, and in activity-dependent adult plasticity. To unravel the role of endogenous neurotrophins in the development of neural connections in the CNS, we studied the ontogeny of hippocampal afferents in trkB (-/-) and trkC (-/-) mice. Injections of lipophilic tracers in the entorhinal cortex and hippocampus of newborn mutant mice showed that the ingrowth of entorhinal and commissural/associational afferents to the hippocampus was not affected by these mutations. Similarly, injections of biocytin in postnatal mutant mice (P10-P16) did not reveal major differences in the topographic patterns of hippocampal connections. In contrast, quantification of biocytin-filled axons showed that commissural and entorhinal afferents have a reduced number of axon collaterals (21-49%) and decreased densities of axonal varicosities (8-17%) in both trkB (-/-) and trkC (-/-) mice. In addition, electron microscopic analyses showed that trkB (-/-) and trkC (-/-) mice have lower densities of synaptic contacts and important structural alterations of presynaptic boutons, such as decreased density of synaptic vesicles. Finally, immunocytochemical studies revealed a reduced expression of the synaptic-associated proteins responsible for synaptic vesicle exocytosis and neurotransmitter release (v-SNAREs and t-SNAREs), especially in trkB (-/-) mice. We conclude that neither trkB nor trkC genes are essential for the ingrowth or layer-specific targeting of hippocampal connections, although the lack of these receptors results in reduced axonal arborization and synaptic density, which indicates a role for TrkB and TrkC receptors in the developmental regulation of synaptic inputs in the CNS in vivo. The data also suggest that the genes encoding for synaptic proteins may be targets of TrkB and TrkC signaling pathways.


Subject(s)
Calcium-Binding Proteins , Hippocampus/cytology , Membrane Proteins , Neuroprotective Agents/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Nerve Growth Factor/genetics , Signal Transduction/physiology , Synapses/physiology , Animals , Antigens, Surface/analysis , Entorhinal Cortex/chemistry , Entorhinal Cortex/cytology , Entorhinal Cortex/embryology , Female , Gene Expression Regulation, Developmental , Hippocampus/chemistry , Hippocampus/embryology , Male , Membrane Glycoproteins/analysis , Mice , Mice, Knockout , Nerve Tissue Proteins/analysis , Neural Pathways , Neurons/chemistry , Neurons/physiology , Neuroprotective Agents/analysis , Organ Culture Techniques , Phenotype , Receptor Protein-Tyrosine Kinases/analysis , Receptor, Ciliary Neurotrophic Factor , Receptor, trkC , Receptors, Nerve Growth Factor/analysis , Synapses/chemistry , Synaptophysin/analysis , Synaptosomal-Associated Protein 25 , Synaptotagmins , Syntaxin 1
10.
Nature ; 385(6611): 70-4, 1997 Jan 02.
Article in English | MEDLINE | ID: mdl-8985248

ABSTRACT

During development of the nervous system, specific recognition molecules provide the cues necessary for the formation of neural connections. In some regions, guiding cues for axonal pathfinding and target selection are provided by specific cells that exist only transiently during development, such as the floorplate or the cortical subplate. In the hippocampus, distinct groups of fibres innervate different layers. We have tested the hypothesis that transient neurons in the hippocampus provide positional information for the targeting of these fibres. Here we report that ablation of Cajal-Retzius cells in organotypic slice cultures of hippocampus prevented the ingrowth of entorhinal but not of commissural afferents. Experiments inhibiting Reelin (an extracellular matrix protein expressed by Cajal-Retzius cells) and analysis of reeler mutant mice showed dramatic abnormalities in the development of entorhinal afferents. Thus Cajal-Retzius cells and reelin are essential for the formation of layer-specific hippocampal connections.


Subject(s)
Astrocytes/physiology , Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Hippocampus/cytology , Neural Pathways/embryology , Animals , Antibodies, Monoclonal , Axons/physiology , Cell Adhesion Molecules, Neuronal/genetics , Culture Techniques , Entorhinal Cortex/cytology , Entorhinal Cortex/embryology , Extracellular Matrix Proteins/genetics , Hippocampus/embryology , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins , Neurons, Afferent/physiology , Reelin Protein , Serine Endopeptidases
11.
J Neurosci ; 16(21): 6896-907, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8824328

ABSTRACT

Cajal-Retzius (CR) cells are transient, pioneer neurons of layer I of the cortex that are believed to play essential roles in corticogenesis, e.g., in neuronal migration and synaptogenesis. Here we have used calretinin immunostaining to study the characteristics, survival, and fate of CR cells in single organotypic slice cultures of mouse neocortex and hippocampus deprived of their extrinsic afferents. In neocortical explants, CR cells were observed after 1-3 d in vitro (DIV), but they disappeared after 5-7 DIV, which is similar to their time of degeneration in vivo. The disappearance of CR cells in neocortical slices was prevented by incubation with tetrodotoxin and the glutamate receptor antagonist 6-cyano-7-nitroquinoxaline-2,3,-dione but not by 2-amino-5-phosphonopentanoic acid, suggesting that neuronal activity and non-NMDA glutamate receptors may trigger CR cell death in the neocortex. In contrast to the situation in vivo, in which many hippocampal CR cells disappear at approximately the third postnatal week, CR cells survived in single hippocampal cultures after long incubation times (31 DIV), with their morphology essentially unaltered. In contrast, fewer CR cells were found when hippocampal slices were cocultured with explants from the entorhinal cortex. Because CR cells are transient synaptic targets for entorhinohippocampal afferents, these findings suggest a role for entorhinal afferents in the degeneration of CR cells in the hippocampus. In conclusion, this study shows different survival properties of CR cells in organotypic slice cultures of hippocampus and neocortex, and it suggests that different mechanisms are involved in the regulation of the process of naturally occurring CR cell death in the two cortical regions.


Subject(s)
Cerebral Cortex/cytology , Hippocampus/cytology , Neurons/cytology , Somatosensory Cortex/cytology , Afferent Pathways , Animals , Animals, Newborn , Cell Death/physiology , Cell Survival/physiology , Cells, Cultured/cytology , Cells, Cultured/ultrastructure , Cerebral Cortex/growth & development , Hippocampus/growth & development , Mice , Mice, Inbred Strains , Microscopy, Electron , Nerve Degeneration/physiology , Neurons/ultrastructure , Organ Culture Techniques , Somatosensory Cortex/growth & development , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...