Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
EMBO Rep ; 21(3): e48512, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31919978

ABSTRACT

Regulation of axon guidance and pruning of inappropriate synapses by class 3 semaphorins are key to the development of neural circuits. Collapsin response mediator protein 2 (CRMP2) has been shown to regulate axon guidance by mediating semaphorin 3A (Sema3A) signaling; however, nothing is known about its role in synapse pruning. Here, using newly generated crmp2-/- mice we demonstrate that CRMP2 has a moderate effect on Sema3A-dependent axon guidance in vivo, and its deficiency leads to a mild defect in axon guidance in peripheral nerves and the corpus callosum. Surprisingly, crmp2-/- mice display prominent defects in stereotyped axon pruning in hippocampus and visual cortex and altered dendritic spine remodeling, which is consistent with impaired Sema3F signaling and with models of autism spectrum disorder (ASD). We demonstrate that CRMP2 mediates Sema3F signaling in primary neurons and that crmp2-/- mice display ASD-related social behavior changes in the early postnatal period as well as in adults. Together, we demonstrate that CRMP2 mediates Sema3F-dependent synapse pruning and its dysfunction shares histological and behavioral features of ASD.


Subject(s)
Autism Spectrum Disorder , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Semaphorins , Animals , Dendritic Spines , Mice , Mice, Knockout , Neuronal Plasticity , Neurons , Signal Transduction
2.
Cell Tissue Res ; 375(1): 23-39, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29869716

ABSTRACT

The neuroendocrine system consists of a heterogeneous collection of (mostly) neuropeptidergic neurons found in four hypothalamic nuclei and sharing the ability to secrete neurohormones (all of them neuropeptides except dopamine) into the bloodstream. There are, however, abundant hypothalamic non-neuroendocrine neuropeptidergic neurons developing in parallel with the neuroendocrine system, so that both cannot be entirely disentangled. This heterogeneity results from the workings of a network of transcription factors many of which are already known. Olig2 and Fezf2 expressed in the progenitors, acting through mantle-expressed Otp and Sim1, Sim2 and Pou3f2 (Brn2), regulate production of magnocellular and anterior parvocellular neurons. Nkx2-1, Rax, Ascl1, Neurog3 and Dbx1 expressed in the progenitors, acting through mantle-expressed Isl1, Dlx1, Gsx1, Bsx, Hmx2/3, Ikzf1, Nr5a2 (LH-1) and Nr5a1 (SF-1) are responsible for tuberal parvocellular (arcuate nucleus) and other neuropeptidergic neurons. The existence of multiple progenitor domains whose progeny undergoes intricate tangential migrations as one source of complexity in the neuropeptidergic hypothalamus is the focus of much attention. How neurosecretory cells target axons to the medial eminence and posterior hypophysis is gradually becoming clear and exciting progress has been made on the mechanisms underlying neurovascular interface formation. While rat neuroanatomy and targeted mutations in mice have yielded fundamental knowledge about the neuroendocrine system in mammals, experiments on chick and zebrafish are providing key information about cellular and molecular mechanisms. Looking forward, data from every source will be necessary to unravel the ways in which the environment affects neuroendocrine development with consequences for adult health and disease.


Subject(s)
Hypothalamus/cytology , Mammals/metabolism , Neurons/cytology , Neurosecretory Systems/cytology , Animals , Cell Movement , Gene Regulatory Networks
3.
Proc Natl Acad Sci U S A ; 111(29): E2996-3004, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002511

ABSTRACT

Celsr3 and Fzd3, members of "core planar cell polarity" (PCP) genes, were shown previously to control forebrain axon guidance and wiring by acting in axons and/or guidepost cells. Here, we show that Celsr2 acts redundantly with Celsr3, and that their combined mutation mimics that of Fzd3. The phenotypes generated upon inactivation of Fzd3 in different forebrain compartments are similar to those in conditional Celsr2-3 mutants, indicating that Fzd3 and Celsr2-3 act in the same population of cells. Inactivation of Celsr2-3 or Fzd3 in thalamus does not affect forebrain wiring, and joint inactivation in cortex and thalamus adds little to cortical inactivation alone in terms of thalamocortical projections. On the other hand, joint inactivation perturbs strongly the formation of the barrel field, which is unaffected upon single cortical or thalamic inactivation, indicating a role for interactions between thalamic axons and cortical neurons in cortical arealization. Unexpectedly, forebrain wiring is normal in mice defective in Vangl1 and Vangl2, showing that, contrary to epithelial PCP, axon guidance can be Vangl independent in some contexts. Our results suggest that Celsr2-3 and Fzd3 regulate axonal navigation in the forebrain by using mechanisms different from classical epithelial PCP, and require interacting partners other than Vangl1-2 that remain to be identified.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Frizzled Receptors/metabolism , Membrane Proteins/metabolism , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Prosencephalon/metabolism , Receptors, Cell Surface/metabolism , Animals , Axons/metabolism , Cerebral Cortex/metabolism , Gene Silencing , Integrases/metabolism , Mice , Mutation/genetics , Phenotype , Thalamus/metabolism
4.
Elife ; 122024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300670

ABSTRACT

Foxb1 -expressing neurons occur in the dorsal premammillary nucleus (PMd) and further rostrally in the parvafox nucleus, a longitudinal cluster of neurons in the lateral hypothalamus of rodents. The descending projection of these Foxb1+ neurons end in the dorsolateral part of the periaqueductal gray (dlPAG). The functional role of the Foxb1+ neuronal subpopulation in the PMd and the parvafox nucleus remains elusive. In this study, the activity of the Foxb1+ neurons and of their terminal endings in the dlPAG in mice was selectively altered by employing chemo- and optogenetic tools. Our results show that in whole-body barometric plethysmography, hM3Dq-mediated, global Foxb1+ neuron excitation activates respiration. Time-resolved optogenetic gain-of-function manipulation of the terminal endings of Foxb1+ neurons in the rostral third of the dlPAG leads to abrupt immobility and bradycardia. Chemogenetic activation of Foxb1+ cell bodies and ChR2-mediated excitation of their axonal endings in the dlPAG led to a phenotypical presentation congruent with a 'freezing-like' situation during innate defensive behavior.


Subject(s)
Bradycardia , Optogenetics , Animals , Mice , Hypothalamus , Neurons , Tachypnea , Forkhead Transcription Factors
5.
Genesis ; 50(1): 67-73, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21898763

ABSTRACT

The hypothalamic mammillary region is critical for spatial memory and vestibular processing. Pitx2 encodes a paired-like transcription factor that is highly expressed in the developing mammillary region and is required for subthalamic nucleus formation. Here we analyzed a loss of function Pitx2-TaulacZ knock-in allele to study the effects of Pitx2 deficiency on neuronal projections in the embryonic mammillary region. Pitx2-expressing neurons contribute axons to principal mammillary, mammillotegmental and mammillotectal tracts. Embryos with Pitx2 deficiency exhibit axonal fibers in the principal mammillary tract that are improperly bundled and disorganized, yet project caudally toward the tectum and tegmentum. Embryos with Nestin-Cre mediated conditional Pitx2 deficiency exhibit truncated mammillothalamic tracts (mtt) that fail to elongate, and reduced Pax6-positive cells at the branching point of the principal mammillary and mtt. These data suggest that Pitx2 mediates cell-autonomous and nonautonomous guidance cues necessary for mammillary collaterals destined to project to the anterior thalamus.


Subject(s)
Alleles , Mammillary Bodies/embryology , Nerve Tissue/metabolism , Animals , Axons/metabolism , Female , Fluorescent Antibody Technique/methods , Genotype , Hypothalamus/metabolism , Integrases/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Male , Mammillary Bodies/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence/methods , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Neurons/metabolism , Tegmentum Mesencephali/embryology , Tegmentum Mesencephali/metabolism , Thalamus/embryology , Thalamus/metabolism
6.
J Chem Neuroanat ; 121: 102089, 2022 04.
Article in English | MEDLINE | ID: mdl-35283254

ABSTRACT

The lateral hypothalamus (LHA) is still a poorly understood brain region. Based on published Dlx and Gad gene expression patterns in the embryonic and adult hypothalamus respectively, three large areas are identified in the LHA. A central tuberal LHA region is already well described as it contains neurons producing the peptides melanin-concentrating hormone or hypocretin. This region is rich in GABAergic neurons and is specified by Dlx gene expression in the rodent embryo. Rostrally and caudally bordering the tuberal LHA, two Dlx-GAD-GABA poor regions are then easily delineated. The three regions show different organizational schema. The tuberal region is reticularly organized, connected with the cerebral cortex and the spinal cord, and its embryonic development occurs along the tractus postopticus. The region anterior to it is associated with the stria medullaris in both embryonic and adult subjects. The posterior LHA region is made of differentiated nuclei and includes the subthalamic nucleus. Therefore, the LHA is divided into three distinct parts: in addition to the well-known tuberal LHA, caudal and anterior LHA regions exist that have specific anatomical and functional characteristics. The hypothalamus is made up of several dozens of nuclei or areas that are more or less well differentiated and whose boundaries and arrangements are drawn differently according to authors and atlases (Allen Institute, 2004; Paxinos and Franklin, 2019; Paxinos and Watson, 2013; Swanson, 2004). The dominant hypothesis for more than 50 years is that these structures are distributed within three antero-posterior areas (anterior, tuberal, posterior) and more or less three longitudinal zones (lateral, medial and periventricular) (Fig. 1). In addition to these regions, several adjacent territories are often associated to the hypothalamus. The preoptic area is functionally related to the hypothalamus, but it is better seen as a telencephalic structure based on developmental data (Croizier et al., 2015; Puelles and Rubenstein, 2015). Lately, the zona incerta and the subthalamic nucleus (STN) have also been associated to the hypothalamus on the basis of their connections and development for the STN (Altman and Bayer, 1986; Barbier and Risold, 2021; Swaab et al., 2021). However, the zona incerta is still included in the 'pre-thalamus' or "ventral thalamus" in the embryo (Puelles and Rubenstein, 2015). Thus, the boundaries of the hypothalamus remain blurred around what we can call a 'core' made of the anterior to posterior regions (Brooks, 1988). In addition, unlike other large brain regions that are characterized early on by a molecular signature, i.e. by the embryonic expression of specific molecular markers, data illustrating the distribution of dozens of transcription factors involved in brain patterning and cell lineage specification confirmed the extremely heterogeneous and mosaic nature of the anterior and posterior regions of the hypothalamus (Alvarez-Bolado, 2019; Puelles et al., 2013; Puelles and Rubenstein, 2015). The rich nuclear organization of the medial and periventricular zones of the hypothalamus is consistent with the mosaic expression of developmental genes. The LHA, however, is often perceived as much more homogeneous in its cytoarchitectural organization. At the same time, there is little information regarding the expression of developmental genes in the anterior and posterior territories of the LHA. Most studies focus on the tuberal LHA which expresses many of these genes. Admittedly, even in the adult hypothalamus, the internal boundaries of the LHA are difficult to identify and the same is true in the embryo. Developmental data alone are insufficient to achieve a better understanding of the LHA anatomical organization and for this region as for medial and periventricular zones, a coherence must be established between development and adult anatomical organization. Among the most useful neurochemical markers to identify large regions of the forebrain, those involved in the identification of GABAergic and glutamatergic neurons have proven to be particularly efficient. Indeed, GABAergic neurons are not ubiquitously distributed. Large regions of the forebrain are rich in such cells, including the basal telencephalon, but others contain few or no GABAergic cells and are rich in glutamatergic neurons instead (for example the dorsal thalamus that is free of GABA-neurons in rodents). The same applies for the hypothalamus: several structures of the hypothalamus are free of GABAergic neurons, as, for example, the mammillary nuclei (Hahn et al., 2019). Recently, we also identified a GABA-poor posterior LHA territory that includes the (STN), and is localized caudal to the GABA-rich tuberal LHA (Barbier et al., 2020; Barbier and Risold, 2021; Chometton et al., 2016b). Therefore, the LHA seems partitioned into GABA-rich/GABA-poor regions. However, to define or confirm distinct neuroanatomical entities, these regions must have a specific embryological origin, and show specific hodological patterns and functions. Hence, the purpose of this short review is to identify divisions of the LHA based on developmental and neurochemical criteria. Such an analysis seems to us relevant in order to allow later functional studies on regions whose boundaries will be based on objective criteria.


Subject(s)
Glutamate Decarboxylase , Rodentia , Animals , Female , Glutamate Decarboxylase/metabolism , Humans , Hypothalamus/metabolism , Pregnancy , Prosencephalon/metabolism , Transcription Factors/metabolism , gamma-Aminobutyric Acid
7.
Transl Psychiatry ; 12(1): 51, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115485

ABSTRACT

Altered long-range connectivity is a common finding across neurodevelopmental psychiatric disorders, but causes and consequences are not well understood. Genetic variation in ST8SIA2 has been associated with schizophrenia, autism, and bipolar disorder, and St8sia2-/- mice show a number of related neurodevelopmental and behavioral phenotypes. In the present study, we use conditional knockout (cKO) to dissect neurodevelopmental defects and behavioral consequences of St8sia2 deficiency in cortical interneurons, their cortical environment, or in the di- and mesencephalon. Neither separate nor combined cortical and diencephalic ablation of St8sia2 caused the disturbed thalamus-cortex connectivity observed in St8sia2-/- mice. However, cortical ablation reproduced hypoplasia of corpus callosum and fornix and mice with di- and mesencephalic ablation displayed smaller mammillary bodies with a prominent loss of parvalbumin-positive projection neurons and size reductions of the mammillothalamic tract. In addition, the mammillotegmental tract and the mammillary peduncle, forming the reciprocal connections between mammillary bodies and Gudden's tegmental nuclei, as well as the size of Gudden's ventral tegmental nucleus were affected. Only mice with these mammillary deficits displayed enhanced MK-801-induced locomotor activity, exacerbated impairment of prepulse inhibition in response to apomorphine, and hypoanxiety in the elevated plus maze. We therefore propose that compromised mammillary body connectivity, independent from hippocampal input, leads to these psychotic-like responses of St8sia2-deficient mice.


Subject(s)
Mammillary Bodies , Sialyltransferases , Animals , Mammillary Bodies/physiology , Mesencephalon , Mice , Tegmentum Mesencephali
8.
Mol Cell Neurosci ; 43(2): 222-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19944164

ABSTRACT

The proliferation, migration and differentiation of dentate gyrus stem and precursor cells have aroused keen interest. Neogenin and RGMb are expressed in non-overlapping compartments of the developing dentate gyrus. While Neogenin is expressed in migrating and proliferating dentate precursors, RGMb is localized in structures bordering the developing dentate, such as cornus ammonis cells and Cajal-Retzius cells in the marginal zone including the hippocampal fissure. Co-immunoprecipitation and binding assays indicate a strong physical interaction. In vitro and in vivo migration of dentate neuroepithelial cells is abolished by RGMb, and cell adhesion is reduced when cells expressing Neogenin comes into contact with cells expressing RGMb. Ectopic expression of RGMb in organotypic slice cultures and after in utero electroporation in the hippocampus modifies precursor cell migration. Our results imply that Neogenin-RGMb interaction might be involved in neuronal migration in the dentate gyrus.


Subject(s)
Cell Movement/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Animals , Cell Adhesion Molecules, Neuronal , Cell Line, Transformed , Electroporation/methods , Embryo, Mammalian , Fluoresceins , GPI-Linked Proteins , Gene Expression Regulation, Developmental/physiology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Homeodomain Proteins/metabolism , Humans , Immunoprecipitation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Organ Culture Techniques , Stem Cells/physiology , Transfection/methods , Tumor Suppressor Proteins/metabolism
9.
Proc Natl Acad Sci U S A ; 105(33): 12016-21, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18687884

ABSTRACT

TRIM RING finger proteins have been shown to play an important role in cancerogenesis, in the pathogenesis of some human hereditary disorders, and in the defense against viral infection, but the function of the majority of TRIM proteins remains unknown. Here, we show that TRIM RING finger protein TRIM2, highly expressed in the nervous system, is an UbcH5a-dependent ubiquitin ligase. We further demonstrate that TRIM2 binds to neurofilament light subunit (NF-L) and regulates NF-L ubiquitination. Additionally, we show that mice deficient in TRIM2 have increased NF-L level in axons and NF-L-filled axonal swellings in cerebellum, retina, spinal cord, and cerebral cortex. The axonopathy is followed by progressive neurodegeneration accompanied by juvenile-onset tremor and ataxia. Our results demonstrate that TRIM2 is an ubiquitin ligase and point to a mechanism regulating NF-L metabolism through an ubiquitination pathway that, if deregulated, triggers neurodegeneration.


Subject(s)
Neurons/metabolism , Proteins/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Gene Expression Regulation , Mice , Microscopy, Immunoelectron , Mutation/genetics , Neurons/ultrastructure , Protein Binding , Proteins/genetics , Time Factors , Tripartite Motif Proteins , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
10.
J Chem Neuroanat ; 116: 101997, 2021 10.
Article in English | MEDLINE | ID: mdl-34182088

ABSTRACT

In the last few years we assist to an unexpected deluge of genomic data on hypothalamic development and structure. Perhaps most surprisingly, the Lateral Zone has received much attention too. The new information focuses first of all on transcriptional heterogeneity. Many already known and a number of hitherto unknown lateral hypothalamic neurons have been described to an enormous degree of detail. Maybe the most surprising novel discoveries are two: First, some restricted regions of the embryonic forebrain neuroepithelium generate specific LHA neurons, either GABAergic or glutamatergic. Second, evidence is mounting that supports the existence of numerous kinds of "bilingual" lateral hypothalamic neurons, expressing (and releasing) glutamate and GABA both as well as assorted neuropeptides. This is not accepted by all, and it could be that genomic researchers need a common set of rules to interpret their data (sensitivity, significance, age of analysis). In any case, some of the new results appear to confirm hypotheses about the ability of the hypothalamus and in particular its Lateral Zone to achieve physiological flexibility on a fixed connectivity ("biochemical switching"). Furthermore, the results succinctly reviewed here are the basis for future advances, since the transcriptional databases generated can now be mined e.g. for adhesion genes, to figure out the causes of the peculiar histology of the Lateral Zone; or for ion channel genes, to clarify present and future electrophysiological data. And with the specific expression data about small subpopulations of neurons, their connections can now be specifically labeled, revealing novel relations with functional significance.


Subject(s)
GABAergic Neurons/chemistry , GABAergic Neurons/metabolism , Glutamic Acid/metabolism , Hypothalamic Area, Lateral/growth & development , Hypothalamic Area, Lateral/metabolism , Neurogenesis/physiology , Animals , Glutamic Acid/analysis , Humans , Hypothalamic Area, Lateral/chemistry , Transcription Factors/analysis , Transcription Factors/biosynthesis
11.
J Neurosci ; 29(8): 2453-66, 2009 Feb 25.
Article in English | MEDLINE | ID: mdl-19244520

ABSTRACT

The specification of the intricate neuronal assemblies that characterize the forebrain is not well understood. The ventral spinal cord is specified through a concentration gradient of Sonic hedgehog (Shh) protein secreted by the notochord. Shh is expressed also in the forebrain neuroepithelium (neural Shh) and the underlying notochord and prechordal plate. Neural Shh is essential for the development of the prethalamus (ventral thalamus), but its effects on the thalamus (dorsal thalamus) are still unclear. We hypothesized that neural Shh would act on a previously regionalized dorsal diencephalic region to promote the emergence of specific thalamic nuclear and histological traits. To find out, we generated a conditional mouse mutant line specifically lacking Shh expression in the diencephalic neuroepithelium. We show that the transcription factor Gbx2, required for thalamic development downstream Shh, is expressed in our mutant in a restricted thalamic region and is necessary and sufficient for the differentiation of the medial and intralaminar thalamic nuclei. In the rest of the thalamus, neural Shh is required to promote neuronal aggregation into nuclei as well as axonal extension. In this way, the individual thalamic nuclei show differential dependence on Shh, Gbx2, or both for their differentiation. Additionally, Gbx2 is required for the survival of thalamic neurons.


Subject(s)
Cell Differentiation/physiology , Hedgehog Proteins/physiology , Neurons/physiology , Thalamus/cytology , Analysis of Variance , Animals , Apoptosis/genetics , Bromodeoxyuridine/metabolism , Cell Differentiation/genetics , Cell Proliferation , Diencephalon/embryology , Diencephalon/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/deficiency , In Situ Nick-End Labeling , Mice , Mice, Transgenic , Mutation/genetics , Nuclear Proteins/metabolism , Polycomb-Group Proteins , Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Thalamus/embryology
12.
J Neurosci ; 29(21): 6989-7002, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19474326

ABSTRACT

The hypothalamus is a region of the diencephalon with particularly complex patterning. Sonic hedgehog (Shh), encoding a protein with key developmental roles, shows a peculiar and dynamic diencephalic expression pattern. Here, we use transgenic strategies and in vitro experiments to test the hypothesis that Shh expressed in the diencephalic neuroepithelium (neural Shh) coordinates tissue growth and patterning in the hypothalamus. Our results show that neural Shh coordinates anteroposterior and dorsoventral patterning in the hypothalamus and in the diencephalon-telencephalon junction. Neural Shh also coordinates mediolateral hypothalamic patterning, since it is necessary for the lateral hypothalamus to attain proper size and is required for the specification of hypocretin/orexin cells. Finally, neural Shh is necessary to maintain expression of differentiation markers including survival factor Foxb1.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental/physiology , Hedgehog Proteins/metabolism , Hypothalamus/cytology , Neuroepithelial Cells/physiology , Signal Transduction/physiology , Age Factors , Analysis of Variance , Animals , Apoptosis/genetics , Apoptosis/physiology , Body Patterning/genetics , Bromodeoxyuridine/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Embryo, Mammalian , Exons/genetics , Forkhead Transcription Factors/genetics , Green Fluorescent Proteins/genetics , Hedgehog Proteins/genetics , Hypothalamus/embryology , In Situ Nick-End Labeling/methods , In Vitro Techniques , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Pathways/embryology , Signal Transduction/genetics , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
13.
PLoS Genet ; 3(10): 1867-83, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17953485

ABSTRACT

Automated in situ hybridization enables the construction of comprehensive atlases of gene expression patterns in mammals. Such atlases can become Web-searchable digital expression maps of individual genes and thus offer an entryway to elucidate genetic interactions and signaling pathways. Towards this end, an atlas housing approximately 1,000 spatial gene expression patterns of the midgestation mouse embryo was generated. Patterns were textually annotated using a controlled vocabulary comprising >90 anatomical features. Hierarchical clustering of annotations was carried out using distance scores calculated from the similarity between pairs of patterns across all anatomical structures. This process ordered hundreds of complex expression patterns into a matrix that reflects the embryonic architecture and the relatedness of patterns of expression. Clustering yielded 12 distinct groups of expression patterns. Because of the similarity of expression patterns within a group, members of each group may be components of regulatory cascades. We focused on the group containing Pax6, an evolutionary conserved transcriptional master mediator of development. Seventeen of the 82 genes in this group showed a change of expression in the developing neocortex of Pax6-deficient embryos. Electromobility shift assays were used to test for the presence of Pax6-paired domain binding sites. This led to the identification of 12 genes not previously known as potential targets of Pax6 regulation. These findings suggest that cluster analysis of annotated gene expression patterns obtained by automated in situ hybridization is a novel approach for identifying components of signaling cascades.


Subject(s)
Gene Expression Regulation , Genetic Techniques , In Situ Hybridization/methods , Animals , Binding Sites , Cluster Analysis , DNA/genetics , Eye Proteins/metabolism , Gene Expression Profiling , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Models, Genetic , Oligonucleotide Array Sequence Analysis , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Time Factors
14.
Front Cell Dev Biol ; 8: 586158, 2020.
Article in English | MEDLINE | ID: mdl-33330463

ABSTRACT

MicroRNAs regulate gene expression at post-transcriptional levels. Some of them appear to regulate brain development and are involved in neurodevelopmental disorders. This has led to the suggestion that the role of microRNAs in neuronal development and function may be more central than previously appreciated. Here, we review the data about miR-9 function to depict the subtlety, complexity, flexibility and limited functional conservation of this essential developmental regulatory system. On this basis we propose that species-specific actions of miR-9 could underlie to a large degree species differences in brain size, shape and function.

15.
Brain Struct Funct ; 224(1): 293-314, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30315416

ABSTRACT

Although connections between the orbitofrontal cortex (OFC)-the seat of high cognitive functions-the lateral hypothalamus and the periaqueductal grey (PAG) have been recognized in the past, the precise targets of the descending fibres have not been identified. In the present study, viral tracer-transport experiments revealed neurons of the lateral (LO) and the ventrolateral (VLO) OFC (homologous to part of Area 13 in primates) to project to a circumscribed region in the ventrolateral hypothalamus, namely, the horizontally oriented, cylindrical parvalbumin- and Foxb1-expressing (parvafox) nucleus. The fine collaterals stem from coarse axons in the internal capsule and form excitatory synapses specifically with neurons of the parvafox nucleus, avoiding the rest of the hypothalamus. In its further caudal course, this contingent of LO/VLO-axons projects collaterals to the Su3- and the PV2 nuclei, which lie ventral to the aqueduct in the (PAG), where the terminals fields overlap those deriving from the parvafox nucleus itself. The targeting of the parvafox nucleus by the LO/VLO-projections, and the overlapping of their terminal fields within the PAG, suggest that the two cerebral sites interact closely. An involvement of this LO/VLO-driven circuit in the somatic manifestation of behavioural events is conceivable.


Subject(s)
Hypothalamic Area, Lateral/physiology , Periaqueductal Gray/physiology , Prefrontal Cortex/physiology , Animals , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genes, Reporter , Hypothalamic Area, Lateral/metabolism , Hypothalamic Area, Lateral/ultrastructure , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/physiology , Neuroanatomical Tract-Tracing Techniques/methods , Parvalbumins/genetics , Parvalbumins/metabolism , Periaqueductal Gray/metabolism , Periaqueductal Gray/ultrastructure , Prefrontal Cortex/metabolism , Prefrontal Cortex/ultrastructure , Rats, Wistar , Recombinant Fusion Proteins/metabolism
16.
Eur J Neurosci ; 28(10): 1941-55, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19046377

ABSTRACT

The hypothalamus is a brain region with vital functions, and alterations in its development can cause human disease. However, we still do not have a complete description of how this complex structure is put together during embryonic and early postnatal stages. Radially oriented, outside-in migration of cells is prevalent in the developing hypothalamus. In spite of this, cell contingents from outside the hypothalamus as well as tangential hypothalamic migrations also have an important role. Here we study migrations in the hypothalamic primordium by genetically labeling the Foxb1 diencephalic lineage. Foxb1 is a transcription factor gene expressed in the neuroepithelium of the developing neural tube with a rostral expression boundary between caudal and rostral diencephalon, and therefore appropriate for marking migrations from caudal levels into the hypothalamus. We have found a large, longitudinally oriented migration stream apparently originating in the thalamic region and following an axonal bundle to end in the anterior portion of the lateral hypothalamic area. Additionally, we have mapped a specific expansion of the neuroepithelium into the rostral diencephalon. The expanded neuroepithelium generates abundant neurons for the medial hypothalamus at the tuberal level. Finally, we have uncovered novel diencephalon-to-telencephalon migrations into septum, piriform cortex and amygdala.


Subject(s)
Cell Lineage/genetics , Cell Movement/genetics , Forkhead Transcription Factors/genetics , Hypothalamus/embryology , Stem Cells/metabolism , Telencephalon/embryology , Animals , Brain Mapping/methods , Diencephalon/embryology , Genetic Markers/genetics , Hypothalamic Area, Lateral/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Biology/methods , Neural Pathways/embryology , Neural Tube/embryology , Stem Cells/cytology
17.
Front Cell Neurosci ; 12: 402, 2018.
Article in English | MEDLINE | ID: mdl-30483057

ABSTRACT

The dentate gyrus (DG) receives highly processed information from the associative cortices functionally integrated in the trisynaptic hippocampal circuit, which contributes to the formation of new episodic memories and the spontaneous exploration of novel environments. Remarkably, the DG is the only brain region currently known to have high rates of neurogenesis in adults (Andersen et al., 1966, 1971). The DG is involved in several neurodegenerative disorders, including clinical dementia, schizophrenia, depression, bipolar disorder and temporal lobe epilepsy. The principal neurons of the DG are the granule cells. DG granule cells generated in culture would be an ideal model to investigate their normal development and the causes of the pathologies in which they are involved and as well as possible therapies. Essential to establish such in vitro models is the precise definition of the most important cell-biological requirements for the differentiation of DG granule cells. This requires a deeper understanding of the precise molecular and functional attributes of the DG granule cells in vivo as well as the DG cells derived in vitro. In this review we outline the neuroanatomical, molecular and cell-biological components of the granule cell differentiation pathway, including some growth- and transcription factors essential for their development. We summarize the functional characteristics of DG granule neurons, including the electrophysiological features of immature and mature granule cells and the axonal pathfinding characteristics of DG neurons. Additionally, we discuss landmark studies on the generation of dorsal telencephalic precursors from pluripotent stem cells (PSCs) as well as DG neuron differentiation in culture. Finally, we provide an outlook and comment critical aspects.

18.
Genesis ; 45(12): 781-7, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18064677

ABSTRACT

We have knocked-in Cre-IRES-EGFP in the Foxb1 locus by homologous recombination in embryonic stem cells. We removed the PGK-neo cassette (which was flanked by FRT sequences) by crossing with the FLPeR deleter mouse. The Foxb1(Cre) line showed Cre recombinase activity as well as EGFP fluorescence reproducing Foxb1 expression accurately. By crossing Foxb1(Cre) mice with the ROSA26R and Z/AP mouse reporter lines we have been able to trace the lineage of Foxb1-expressing cells. Early transient expression of Foxb1 in the paraxial mesoderm translates into labeling of the somites. In the central nervous system (CNS), the Foxb1 lineage includes the thalamus and mammillary body (hypothalamus), brainstem, and the ventral spinal cord and floor plate.


Subject(s)
Central Nervous System/embryology , Forkhead Transcription Factors/genetics , Integrases/genetics , Mutagenesis, Insertional/methods , Animals , Brain Stem/embryology , Brain Stem/metabolism , Central Nervous System/metabolism , Diencephalon/embryology , Diencephalon/metabolism , Embryonic Stem Cells/metabolism , Green Fluorescent Proteins/genetics , Integrases/analysis , Integrases/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , Recombination, Genetic , Somites/embryology , Somites/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism
19.
Front Neuroanat ; 11: 53, 2017.
Article in English | MEDLINE | ID: mdl-28725186

ABSTRACT

Oligodendrocyte precursor cells (OPC), neurons and astrocytes share a neural progenitor cell (NPC) in the early ventricular zone (VZ) of the embryonic neuroepithelium. Both switch to produce either of the three cell types and the generation of the right number of them undergo complex genetic regulation. The components of these regulatory cascades vary between brain regions giving rise to the unique morphological and functional heterogeneity of this organ. Forkhead b1 (Foxb1) is a transcription factor gene expressed by NPCs in specific regions of the embryonic neuroepithelium. We used the mutant mouse line Foxb1-Cre to analyze the cell types derived from Fobx1-expressing NPCs (the Foxb1 cell lineage) from two restricted regions, the medulla oblongata (MO; hindbrain) and the thalamus (forebrain), of normal and Foxb1-deficient mice. Foxb1 cell lineage derivatives appear as clusters in restricted regions, including the MO (hindbrain) and the thalamus (forebrain). Foxb1-expressing NPCs produce mostly oligodendrocytes (OL), some neurons and few astrocytes. Foxb1-deficient NPCs generate mostly OPC and immature OL to the detriment of neurons, astrocytes and mature OL. The axonal G-ratio however is not changed. We reveal Foxb1 as a novel modulator of neuronal and OL generation in certain restricted CNS regions. Foxb1 biases NPCs towards neuronal generation and inhibits OPC proliferation while promoting their differentiation.

20.
J Comp Neurol ; 496(5): 684-97, 2006 Jun 10.
Article in English | MEDLINE | ID: mdl-16615126

ABSTRACT

Adenylate cyclases (Adcys) are components of several developmentally, neurophysiologically, and pharmacologically relevant signaling pathways. A prominent feature of Adcys is their ability to integrate multiple signaling pathways into a single second messenger pathway, the production of cAMP. Nine isoforms of membrane-bound Adcys are known, each encoded by a distinct gene. These isoforms differ in their response to regulatory upstream pathways as well as in their distribution in the brain and elsewhere. Use of various detection methods and animal species has, however, hampered a direct comparison of expression patterns, so the potential contribution of single isoforms to Adcy activity in different brain regions remains unclear. We have determined the expression patterns of all nine Adcy genes in the embryonic, postnatal day 7, and adult mouse brain by nonradioactive robotic in situ hybridization (ISH). Here we describe the salient features of these patterns. Regional colocalization of Adcy transcripts encoding isoforms with different regulatory properties was detected in the cortex, subregions of the hippocampus, olfactory bulb, thalamus, and striatum. Hence, our expression data support models for modulation of cAMP signaling by combinatorial action of multiple Adcy isoforms. However, in several instances, the expression domains of genes encoding isoforms with similar regulatory properties spatially exclude each other, which is most evident in not previously described expression domains of the embryonic midbrain roof. This is suggestive of functional specialization.


Subject(s)
Adenylyl Cyclases/biosynthesis , Brain/enzymology , Animals , Animals, Newborn , Brain/embryology , Brain/growth & development , Female , In Situ Hybridization , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , Robotics
SELECTION OF CITATIONS
SEARCH DETAIL