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










Publication year range
1.
Int J Mol Sci ; 22(20)2021 Oct 17.
Article in English | MEDLINE | ID: mdl-34681849

ABSTRACT

We generated and characterized a transgenic mouse line with the tendon-specific expression of a double fluorescent reporter system, which will fulfill an unmet need for animal models to support real-time monitoring cell behaviors during tendon development, growth, and repair in vitro and in vivo. The mScarlet red fluorescent protein is driven by the Scleraxis (Scx) promoter to report the cell lineage alteration. The blue fluorescent protein reporter is expressed under the control of the 3.6kb Collagen Type I Alpha 1 Chain (Col1a1) proximal promoter. In this promoter, the existence of two promoter regions named tendon-specific cis-acting elements (TSE1, TSE2) ensure the specific expression of blue fluorescent protein (BFP) in tendon tissue. Collagen I is a crucial marker for tendon regeneration that is a major component of healthy tendons. Thus, the alteration of function during tendon repair can be estimated by BFP expression. After mechanical stimulation, the expression of mScarlet and BFP increased in adipose-derived mesenchymal stem cells (ADMSCs) from our transgenic mouse line, and there was a rising trend on tendon key markers. These results suggest that our tendon-specific double reporter system is a novel model used to study cell re-differentiation and extracellular matrix alteration in vitro and in vivo.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Collagen Type I, alpha 1 Chain/genetics , Mice, Transgenic , Promoter Regions, Genetic , Tendons/metabolism , Animals , Cell Tracking , Mesenchymal Stem Cells , Mice , Tendons/growth & development , Tendons/physiology
2.
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
3.
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
4.
Sci Rep ; 9(1): 16173, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31700011

ABSTRACT

Aberrant histone acetylation contributes to age-dependent cognitive decline and neurodegenerative diseases. We analyze the function of lysine acetyltransferase TIP60/KAT5 in neurons of the hippocampus using an inducible mouse model. TIP60-deficiency in the adult forebrain leads within days to extensive transcriptional dysfunction characterized by the presence of a neurodegeneration-related signature in CA1. Cell cycle- and immunity-related genes are upregulated while learning- and neuronal plasticity-related genes are downregulated. The dysregulated genes seen under TIP60-deficiency overlap with those in the well-characterized CK-p25 neurodegeneration model. We found that H4K12 is hypoacetylated at the transcriptional start sites of those genes whose expression is dampened in TIP60-deficient mice. Transcriptional dysregulation is followed over a period of weeks by activation of Caspase 3 and fragmentation of ß-actin in CA1 neurites, eventually leading to severe neuronal loss. TIP60-deficient mice also develop mild memory impairment. These phenotypes point to a central role of TIP60 in transcriptional networks that are critical for neuronal viability.


Subject(s)
CA1 Region, Hippocampal/metabolism , Lysine Acetyltransferase 5/metabolism , Memory Disorders/metabolism , Neurites/metabolism , Neurodegenerative Diseases/metabolism , Trans-Activators/metabolism , Acetylation , Animals , CA1 Region, Hippocampal/pathology , Cell Survival/genetics , Lysine Acetyltransferase 5/genetics , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Mice, Transgenic , Neurites/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Trans-Activators/genetics
5.
Elife ; 82019 07 11.
Article in English | MEDLINE | ID: mdl-31294688

ABSTRACT

Many physiological processes exhibit circadian rhythms driven by cellular clocks composed of interlinked activating and repressing elements. To investigate temporal regulation in this molecular oscillator, we combined mouse genetic approaches and analyses of interactions of key circadian proteins with each other and with clock gene promoters. We show that transcriptional activators control BRD4-PTEFb recruitment to E-box-containing circadian promoters. During the activating phase of the circadian cycle, the lysine acetyltransferase TIP60 acetylates the transcriptional activator BMAL1 leading to recruitment of BRD4 and the pause release factor P-TEFb, followed by productive elongation of circadian transcripts. We propose that the control of BRD4-P-TEFb recruitment is a novel temporal checkpoint in the circadian clock cycle.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Rhythm/genetics , Lysine Acetyltransferase 5/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Basic-Leucine Zipper Transcription Factors/genetics , CLOCK Proteins/genetics , Circadian Clocks/genetics , E-Box Elements/genetics , Mice , Promoter Regions, Genetic , Protein Binding/genetics , Transcriptional Activation/genetics
6.
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.

7.
Front Neuroanat ; 9: 113, 2015.
Article in English | MEDLINE | ID: mdl-26321924

ABSTRACT

Acquisition of specific neuronal identity by individual brain nuclei is a key step in brain development. However, how the mechanisms that confer neuronal identity are integrated with upstream regional specification networks is still mysterious. Expression of Sonic hedgehog (Shh), is required for hypothalamic specification and is later downregulated by Tbx3 to allow for the differentiation of the tubero-mamillary region. In this region, the mamillary body (MBO), is a large neuronal aggregate essential for memory formation. To clarify how MBO identity is acquired after regional specification, we investigated Lhx5, a transcription factor with restricted MBO expression. We first generated a hypomorph allele of Lhx5-in homozygotes, the MBO disappears after initial specification. Intriguingly, in these mutants, Tbx3 was downregulated and the Shh expression domain abnormally extended. Microarray analysis and chromatin immunoprecipitation indicated that Lhx5 appears to be involved in Shh downregulation through Tbx3 and activates several MBO-specific regulator and effector genes. Finally, by tracing the caudal hypothalamic cell lineage we show that, in the Lhx5 mutant, at least some MBO cells are present but lack characteristic marker expression. Our work shows how the Lhx5 locus contributes to integrate regional specification pathways with downstream acquisition of neuronal identity in the MBO.

9.
Front Neuroanat ; 9: 29, 2015.
Article in English | MEDLINE | ID: mdl-25852491

ABSTRACT

Expression of intricate combinations of cadherins (a family of adhesive membrane proteins) is common in the developing central nervous system. On this basis, a combinatorial cadherin code has long been proposed to underlie neuronal sorting and to be ultimately responsible for the layers, columns and nuclei of the brain. However, experimental proof of this particular function of cadherins has proven difficult to obtain and the question is still not clear. Alternatively, non-specific, non-combinatorial, purely quantitative adhesive differentials have been proposed to explain neuronal sorting in the brain. Do cadherin combinations underlie brain cytoarchitecture? We approached this question using as model a well-defined forebrain nucleus, the mammillary body (MBO), which shows strong, homogeneous expression of one single cadherin (Cdh11) and patterned, combinatorial expression of Cdh6, -8 and -10. We found that, besides the known combinatorial Cdh pattern, MBO cells are organized into a second, non-overlapping pattern grouping neurons with the same date of neurogenesis. We report that, in the Foxb1 mouse mutant, Cdh11 expression fails to be maintained during MBO development. This disrupted the combination-based as well as the birthdate-based sorting in the mutant MBO. In utero RNA interference (RNAi) experiments knocking down Cdh11 in MBO-fated migrating neurons at one specific age showed that Cdh11 expression is required for chronological entrance in the MBO. Our results suggest that neuronal sorting in the developing MBO is caused by adhesion-based, non-combinatorial mechanisms that keep neurons sorted according to birthdate information (possibly matching them to target neurons chronologically sorted in the same manner). Non-specific adhesion mechanisms would also prevent cadherin combinations from altering the birthdate-based sorting. Cadherin combinations would presumably act later to support specific synaptogenesis through specific axonal fasciculation and final target recognition.

10.
Front Neuroanat ; 9: 34, 2015.
Article in English | MEDLINE | ID: mdl-25859185

ABSTRACT

Secreted protein Sonic hedgehog (Shh) ventralizes the neural tube by modulating the crucial balance between activating and repressing functions (GliA, GliR) of transcription factors Gli2 and Gli3. This balance-the Shh-Gli code-is species- and context-dependent and has been elucidated for the mouse spinal cord. The hypothalamus, a forebrain region regulating vital functions like homeostasis and hormone secretion, shows dynamic and intricate Shh expression as well as complex regional differentiation. Here we asked if particular combinations of Gli2 and Gli3 and of GliA and GliR functions contribute to the variety of hypothalamic regions, i.e., we wanted to approach the question of a possible hypothalamic version of the Shh-Gli code. Based on mouse mutant analysis, we show that: (1) hypothalamic regional heterogeneity is based in part on differentially stringent requirements for Gli2 or Gli3; (2) another source of diversity are differential requirements for Shh of neural vs. non-neural origin; (3) the medial progenitor domain known to depend on Gli2 for its development generates several essential hypothalamic nuclei plus the pituitary and median eminence; (4) the suppression of Gli3R by neural and non-neural Shh is essential for hypothalamic specification. Finally, we have mapped our results on a recent model which considers the hypothalamus as a transverse region with alar and basal portions. Our data confirm the model and are explained by it.

11.
Front Neuroanat ; 8: 156, 2014.
Article in English | MEDLINE | ID: mdl-25610374

ABSTRACT

The expression pattern of Sonic Hedgehog (Shh) in the developing hypothalamus changes over time. Shh is initially expressed in the prechordal mesoderm and later in the hypothalamic neuroepithelium-first medially, and then in two off-medial domains. This dynamic expression suggests that Shh might regulate several aspects of hypothalamic development. To gain insight into them, lineage tracing, (conditional) gene inactivation in mouse, in ovo loss- and gain-of-function approaches in chick and analysis of Shh expression regulation have been employed. We will focus on mouse studies and refer to chick and fish when appropriate to clarify. These studies show that Shh-expressing neuroepithelial cells serve as a signaling center for neighboring precursors, and give rise to most of the basal hypothalamus (tuberal and mammillary regions). Shh signaling is initially essential for hypothalamic induction. Later, Shh signaling from the neuroepithelium controls specification of the lateral hypothalamic area and growth-patterning coordination in the basal hypothalamus. To further elucidate the role of Shh in hypothalamic development, it will be essential to understand how Shh regulates the downstream Gli transcription factors.

12.
J Vis Exp ; (77)2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23912701

ABSTRACT

Genetic modification of specific regions of the developing mammalian brain is a very powerful experimental approach. However, generating novel mouse mutants is often frustratingly slow. It has been shown that access to the mouse brain developing in utero with reasonable post-operatory survival is possible. Still, results with this procedure have been reported almost exclusively for the most superficial and easily accessible part of the developing brain, i.e. the cortex. The thalamus, a narrower and more medial region, has proven more difficult to target. Transfection into deeper nuclei, especially those of the hypothalamus, is perhaps the most challenging and therefore very few results have been reported. Here we demonstrate a procedure to target the entire hypothalamic neuroepithelium or part of it (hypothalamic regions) for transfection through electroporation. The keys to our approach are longer narcosis times, injection in the third ventricle, and appropriate kind and positioning of the electrodes. Additionally, we show results of targeting and subsequent histological analysis of the most recessed hypothalamic nucleus, the mammillary body.


Subject(s)
Electroporation/methods , Hypothalamus/embryology , Hypothalamus/physiology , Animals , DNA/administration & dosage , DNA/genetics , Embryo, Mammalian , Female , Mice , Pregnancy , Transfection/methods
13.
Front Neurosci ; 6: 27, 2012.
Article in English | MEDLINE | ID: mdl-22371696

ABSTRACT

Sonic hedgehog (Shh) signaling is essential for thalamic development. The Gli transcription factors act downstream of Shh - while Gli2 is the major activator (GliA), Gli3 acts primarily as a repressor (GliR). The thalamus is remarkable among dorsal structures because of its proximity to the mid-diencephalic organizer, a unique dorsal Shh source. This lends complexity to the interactions between Shh, Gli2, and Gli3, suggesting the presence of a dorsal Gli activator which elsewhere is found only ventrally, and making the dissection of thalamic Gli functions particularly interesting. A current model based on mutant phenotypes in telencephalon and midbrain postulates a degree of reciprocal antagonism of Shh and Gli3 in dorsal brain regions. To approach the role of Gli factors in thalamic specification we first analyzed mice deficient in Gli2 or Gli3. In Gli2 mutants, the thalamus is small and poorly differentiated with the exception of the medial and intralaminar nuclei which, in contrast, are specifically and severely affected by Gli3 inactivation. Gbx2 expression is very reduced in the Gli3 mutant. Most thalamic nuclei are present in both mutants, although incompletely differentiated, as reflected by the loss of specific markers. The ventral posterior group, revealed by novel specific marker Hes1, is present in both mutants and extends axons to the telencephalon. To test the Gli3/Shh interaction we generated a novel mutant deficient in Gli3 and neuroepithelial Shh. The thalamus of the n-Shh/Gli3 double mutants is very large and very poorly differentiated except for a broad domain of Gbx2, Lhx2, and Calb2 expression. In utero electroporation experiments on wild type embryos suggest that a stage-specific factor acting early is responsible for this prepattern. We show that, in the thalamus, GliA acts downstream of Shh to specify pattern and size of the thalamic nuclei to the exception of the medial and intralaminar groups. Gli3A can partially substitute for Gli2A in the Gli2 mutant. GliR is essential for specification and growth of the medial and intralaminar nuclei, contributes to the specification of other thalamic nuclei and reduces thalamic size. GliA (from neuroepithelial Shh signaling) and GliR do not show reciprocal antagonism in the thalamus, and their joint abolition does not rescue the wild type phenotype.

14.
J Biol Rhythms ; 26(5): 379-89, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21921292

ABSTRACT

Surgical lesion of the suprachiasmatic nuclei (SCN) profoundly affects the circadian timing system. A complication of SCN ablations is the concomitant scission of SCN afferents and efferents. Genetic disruption of the molecular clockwork in the SCN provides a complementary, less invasive experimental approach. The authors report the generation and functional analysis of a new Cre recombinase driver mouse that evokes homologous recombination with high efficiency in the SCN. They inserted the Cre recombinase cDNA into the Synaptotagmin10 (Syt10) locus, a gene strongly expressed in the SCN. Heterozygous Synaptotagmin10-Cre (Syt10(Cre)) mice have no obvious circadian locomotor phenotype, and homozygous animals show slightly reduced light-induced phase delays. Crosses of Syt10(Cre) mice with ß-galactosidase reporter animals revealed strong Cre activity in the vast majority of SCN cells. Cre activity is not detected in nonneuronal tissues with the exception of the testis. The authors demonstrate that conditionally deleting the clock gene Bmal1 using the Syt10(Cre) driver renders animals arrhythmic.


Subject(s)
CLOCK Proteins/genetics , Circadian Rhythm/genetics , Integrases/genetics , Suprachiasmatic Nucleus/physiology , Synaptotagmins/physiology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/physiology , Animals , Behavior, Animal , Circadian Rhythm/physiology , Gene Knock-In Techniques , Homologous Recombination , Male , Mice , Motor Activity/genetics , Synaptotagmins/genetics , beta-Galactosidase/genetics
15.
PLoS One ; 6(5): e20315, 2011.
Article in English | MEDLINE | ID: mdl-21625468

ABSTRACT

BACKGROUND: An essential phenomenon during brain development is the extension of long collateral branches by axons. How the local cellular environment contributes to the initial sprouting of these branches in specific points of an axonal shaft remains unclear. METHODOLOGY/PRINCIPAL FINDINGS: The principal mammillary tract (pm) is a landmark axonal bundle connecting ventral diencephalon to brainstem (through the mammillotegmental tract, mtg). Late in development, the axons of the principal mammillary tract sprout collateral branches at a very specific point forming a large bundle whose target is the thalamus. Inspection of this model showed a number of distinct, identified cell populations originated in the dorsal and the ventral diencephalon and migrating during development to arrange themselves into several discrete groups around the branching point. Further analysis of this system in several mouse lines carrying mutant alleles of genes expressed in defined subpopulations (including Pax6, Foxb1, Lrp6 and Gbx2) together with the use of an unambiguous genetic marker of mammillary axons revealed: 1) a specific group of Pax6-expressing cells in close apposition with the prospective branching point is indispensable to elicit axonal branching in this system; and 2) cooperation of transcription factors Foxb1 and Pax6 to differentially regulate navigation and fasciculation of distinct branches of the principal mammillary tract. CONCLUSIONS/SIGNIFICANCE: Our results define for the first time a model system where interaction of the axonal shaft with a specific group of surrounding cells is essential to promote branching. Additionally, we provide insight on the cooperative transcriptional regulation necessary to promote and organize an intricate axonal tree.


Subject(s)
Axons , Mammillary Bodies/physiology , Animals , Eye Proteins/genetics , Gene Expression Profiling , Homeodomain Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Mutant Strains , Microscopy, Confocal , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics
16.
Dis Model Mech ; 2(7-8): 412-8, 2009.
Article in English | MEDLINE | ID: mdl-19535498

ABSTRACT

Polycomb proteins are epigenetic regulators of gene expression. Human central nervous system (CNS) malformations are congenital defects of the brain and spinal cord. One example of a human CNS malformation is Chiari malformation (CM), which presents as abnormal brainstem growth and cerebellar herniation, sometimes accompanied by spina bifida and cortical defects; it can occur in families. Clinically, CM ranges from an asymptomatic condition to one with incapacitating or lethal symptoms, including neural tube defects and hydrocephalus. However, no genes that are causally involved in any manifestation of CM or similar malformations have been identified. Here, we show that a pathway that involves Zac1 (also known as Plagl1 or Lot1) and controls neuronal proliferation is altered in mice that are heterozygous for the polycomb gene Suz12, resulting in a phenotype that overlaps with some clinical manifestations of the CM spectrum. Suz12 heterozygotes show cerebellar herniation and an enlarged brainstem, accompanied by occipital cortical alterations and spina bifida. Downward displacement of the cerebellum causes hydrocephalus in the most severely impaired cases. Although the involvement of polycomb genes in human disease is starting to be recognized, this is the first demonstration of their role in nervous system malformations. Our work strongly suggests that brain malformations such as CM can result from altered epigenetic regulation of genes involved in cell proliferation in the brain.


Subject(s)
Brain/abnormalities , Brain/metabolism , Gene Expression Regulation, Developmental , Neural Tube Defects/genetics , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Cell Proliferation , Epigenesis, Genetic , Heterozygote , In Situ Hybridization , Mice , Models, Genetic , Phenotype , Polycomb Repressive Complex 2 , Time Factors
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL