Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 149(14)2022 07 15.
Article in English | MEDLINE | ID: mdl-35815619

ABSTRACT

The midbrain reticular formation (MRF) is a mosaic of diverse GABAergic and glutamatergic neurons that have been associated with a variety of functions, including sleep regulation. However, the molecular characteristics and development of MRF neurons are poorly understood. As the transcription factor, Gata2 is required for the development of all GABAergic neurons derived from the embryonic mouse midbrain, we hypothesized that the genes expressed downstream of Gata2 could contribute to the diversification of GABAergic neuron subtypes in this brain region. Here, we show that Gata2 is required for the expression of several GABAergic lineage-specific transcription factors, including Nkx2-2 and Skor2, which are co-expressed in a restricted group of post-mitotic GABAergic precursors in the MRF. Both Gata2 and Nkx2-2 function is required for Skor2 expression in GABAergic precursors. In the adult mouse and rat midbrain, Nkx2-2-and Skor2-expressing GABAergic neurons locate at the boundary of the ventrolateral periaqueductal gray and the MRF, an area containing REM-off neurons regulating REM sleep. In addition to the characteristic localization, Skor2+ cells increase their activity upon REM-sleep inhibition, send projections to the dorsolateral pons, a region associated with sleep control, and are responsive to orexins, consistent with the known properties of midbrain REM-off neurons.


Subject(s)
GABAergic Neurons , Sleep, REM , Animals , GABAergic Neurons/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Homeobox Protein Nkx-2.2/metabolism , Mesencephalon , Mice , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Rats , Sleep/physiology , Sleep, REM/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
2.
J Neurosci ; 40(45): 8669-8682, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33046550

ABSTRACT

Serotonergic neurons in the dorsal raphe (DR) nucleus are associated with several psychiatric disorders including depression and anxiety disorders, which often have a neurodevelopmental component. During embryonic development, GATA transcription factors GATA2 and GATA3 operate as serotonergic neuron fate selectors and regulate the differentiation of serotonergic neuron subtypes of DR. Here, we analyzed the requirement of GATA cofactor ZFPM1 in the development of serotonergic neurons using Zfpm1 conditional mouse mutants. Our results demonstrated that, unlike the GATA factors, ZFPM1 is not essential for the early differentiation of serotonergic precursors in the embryonic rhombomere 1. In contrast, in perinatal and adult male and female Zfpm1 mutants, a lateral subpopulation of DR neurons (ventrolateral part of the DR) was lost, whereas the number of serotonergic neurons in a medial subpopulation (dorsal region of the medial DR) had increased. Additionally, adult male and female Zfpm1 mutants had reduced serotonin concentration in rostral brain areas and displayed increased anxiety-like behavior. Interestingly, female Zfpm1 mutant mice showed elevated contextual fear memory that was abolished with chronic fluoxetine treatment. Altogether, these results demonstrate the importance of ZFPM1 for the development of DR serotonergic neuron subtypes involved in mood regulation. It also suggests that the neuronal fate selector function of GATAs is modulated by their cofactors to refine the differentiation of neuronal subtypes.SIGNIFICANCE STATEMENT Predisposition to anxiety disorders has both a neurodevelopmental and a genetic basis. One of the brainstem nuclei involved in the regulation of anxiety is the dorsal raphe, which contains different subtypes of serotonergic neurons. We show that inactivation of a transcriptional cofactor ZFPM1 in mice results in a developmental failure of laterally located dorsal raphe serotonergic neurons and changes in serotonergic innervation of rostral brain regions. This leads to elevated anxiety-like behavior and contextual fear memory, alleviated by chronic fluoxetine treatment. Our work contributes to understanding the neurodevelopmental mechanisms that may be disturbed in the anxiety disorder.


Subject(s)
Anxiety/genetics , Anxiety/psychology , Dorsal Raphe Nucleus/growth & development , GATA Transcription Factors/genetics , Serotonergic Neurons , Transcription Factors/genetics , Animals , Behavior, Animal , Brain Chemistry/genetics , Dorsal Raphe Nucleus/cytology , Fear/psychology , Female , Fluoxetine/pharmacology , Male , Memory , Mice , Mice, Knockout , Mutation/genetics , Pregnancy , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
3.
Development ; 143(23): 4495-4508, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27789623

ABSTRACT

Serotonergic and glutamatergic neurons of the dorsal raphe regulate many brain functions and are important for mental health. Their functional diversity is based on molecularly distinct subtypes; however, the development of this heterogeneity is poorly understood. We show that the ventral neuroepithelium of mouse anterior hindbrain is divided into specific subdomains giving rise to serotonergic neurons as well as other types of neurons and glia. The newly born serotonergic precursors are segregated into distinct subpopulations expressing vesicular glutamate transporter 3 (Vglut3) or serotonin transporter (Sert). These populations differ in their requirements for transcription factors Gata2 and Gata3, which are activated in the post-mitotic precursors. Gata2 operates upstream of Gata3 as a cell fate selector in both populations, whereas Gata3 is important for the differentiation of the Sert+ precursors and for the serotonergic identity of the Vglut3+ precursors. Similar to the serotonergic neurons, the Vglut3-expressing glutamatergic neurons, located in the central dorsal raphe, are derived from neural progenitors in the ventral hindbrain and express Pet1 Furthermore, both Gata2 and Gata3 are redundantly required for their differentiation. Our study demonstrates lineage relationships of the dorsal raphe neurons and suggests that functionally significant heterogeneity of these neurons is established early during their differentiation.


Subject(s)
Dorsal Raphe Nucleus/cytology , GATA2 Transcription Factor/genetics , GATA3 Transcription Factor/genetics , Neurogenesis/genetics , Rhombencephalon/embryology , Serotonergic Neurons/cytology , Amino Acid Transport Systems, Acidic/metabolism , Animals , Mice , Mice, Knockout , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuroglia/cytology , Rhombencephalon/physiology , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacology , Transcription Factors/biosynthesis
4.
Development ; 143(3): 516-29, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26718003

ABSTRACT

Local inhibitory GABAergic and excitatory glutamatergic neurons are important for midbrain dopaminergic and hindbrain serotonergic pathways controlling motivation, mood, and voluntary movements. Such neurons reside both within the dopaminergic nuclei, and in adjacent brain structures, including the rostromedial and laterodorsal tegmental nuclei. Compared with the monoaminergic neurons, the development, heterogeneity, and molecular characteristics of these regulatory neurons are poorly understood. We show here that different GABAergic and glutamatergic subgroups associated with the monoaminergic nuclei express specific transcription factors. These neurons share common origins in the ventrolateral rhombomere 1, where the postmitotic selector genes Tal1, Gata2 and Gata3 control the balance between the generation of inhibitory and excitatory neurons. In the absence of Tal1, or both Gata2 and Gata3, the GABAergic precursors adopt glutamatergic fates and populate the glutamatergic nuclei in excessive numbers. Together, our results uncover developmental regulatory mechanisms, molecular characteristics, and heterogeneity of central regulators of monoaminergic circuits.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , Dopaminergic Neurons/cytology , Mesencephalon/cytology , Neural Inhibition , Animals , Biomarkers/metabolism , Chickens , Embryo, Mammalian/metabolism , Female , Forkhead Transcription Factors/metabolism , GABAergic Neurons/cytology , GATA Transcription Factors/metabolism , Glutamates/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Mitosis , Models, Biological , Repressor Proteins/metabolism , Serotonin/metabolism , Substantia Nigra/cytology , Ventral Tegmental Area/cytology , gamma-Aminobutyric Acid/metabolism
5.
Am J Med Genet A ; 170(8): 2111-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27183861

ABSTRACT

Clinical classification of overgrowth syndromes represents a challenge since a wide spectrum of disorders result in marked overgrowth. Therefore, there is a continuous effort to identify the genetic basis of these disorders that will eventually facilitate their molecular classification. Here, we have identified the genetic etiology and the pathogenetic mechanism underlying a rare autosomal recessive overgrowth syndrome in three affected siblings. The overgrowth phenotype in the patients was accompanied by developmental delay, learning disabilities, and variable congenital abnormalities. To elucidate the genetic etiology of the disorder, whole-genome genotyping and whole-exome sequencing were used. The disease was mapped to 3p21.1-p14.2 and 11q13.1-q13.4, where an in-frame insertion (c.175_176insTAA) in FIBP gene was revealed. The resulting indel (p.H59LN) was predicted to change the protein conformation with likely deleterious effect on its function as one of the fibroblast growth factor signaling mediators. In vitro cellular proliferation assay and in situ hypridization in vivo were then performed to understand the pathophysiology of the disease. The patients' skin fibroblasts showed an increased proliferation capacity compared to the controls' explaining the observed overgrowth phenotype. In addition, we detected Fibp expression most notably in the brains of mice embryos suggesting a possible effect on cognitive functions early in development. To date, only one patient has been reported with a homozygous nonsense mutation in FIBP exhibiting an overgrowth syndrome with multiple congenital abnormalities. Taken all together, these findings provide convincing evidence implicating FIBP aberrations in the newly recognized overgrowth syndrome and expand the associated phenotypes to include possible Wilms tumor predisposition. © 2016 Wiley Periodicals, Inc.


Subject(s)
Carrier Proteins/genetics , Genes, Recessive , Growth Disorders/genetics , Intellectual Disability/genetics , Kidney/abnormalities , Membrane Proteins/genetics , Mutation , Wilms Tumor/etiology , Adolescent , Animals , Cell Proliferation , Child , Child, Preschool , Chromosome Mapping , DNA Mutational Analysis , Exome , Female , Gene Expression , Gene Expression Regulation, Developmental , Genetic Association Studies , Genotype , Growth Disorders/diagnosis , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Intellectual Disability/diagnosis , Male , Mice , Mice, Transgenic , Pedigree , Phenotype , Syndrome , Wilms Tumor/diagnosis
6.
Development ; 139(5): 894-905, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22278924

ABSTRACT

The structure and projection patterns of adult mesodiencephalic dopaminergic (DA) neurons are one of the best characterized systems in the vertebrate brain. However, the early organization and development of these nuclei remain poorly understood. The induction of midbrain DA neurons requires sonic hedgehog (Shh) from the floor plate and fibroblast growth factor 8 (FGF8) from the isthmic organizer, but the way in which FGF8 regulates DA neuron development is unclear. We show that, during early embryogenesis, mesodiencephalic neurons consist of two distinct populations: a diencephalic domain, which is probably independent of isthmic FGFs; and a midbrain domain, which is dependent on FGFs. Within these domains, DA progenitors and precursors use partly different genetic programs. Furthermore, the diencephalic DA domain forms a distinct cell population, which also contains non-DA Pou4f1(+) cells. FGF signaling operates in proliferative midbrain DA progenitors, but is absent in postmitotic DA precursors. The loss of FGFR1/2-mediated signaling results in a maturation failure of the midbrain DA neurons and altered patterning of the midbrain floor. In FGFR mutants, the DA domain adopts characteristics that are typical for embryonic diencephalon, including the presence of Pou4f1(+) cells among TH(+) cells, and downregulation of genes typical of midbrain DA precursors. Finally, analyses of chimeric embryos indicate that FGF signaling regulates the development of the ventral midbrain cell autonomously.


Subject(s)
Cell Differentiation/physiology , Diencephalon , Dopaminergic Neurons/physiology , Fibroblast Growth Factors/metabolism , Mesencephalon , Morphogenesis/physiology , Signal Transduction/physiology , Animals , Diencephalon/cytology , Diencephalon/embryology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/drug effects , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mesencephalon/cytology , Mesencephalon/embryology , Mice , Pregnancy , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Stem Cells/cytology , Stem Cells/physiology , Tretinoin/pharmacology
7.
Development ; 139(13): 2360-70, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22627282

ABSTRACT

GABAergic neurons in the ventral mesodiencephalic region are highly important for the function of dopaminergic pathways that regulate multiple aspects of behavior. However, development of these neurons is poorly understood. We recently showed that molecular regulation of differentiation of the GABAergic neurons associated with the dopaminergic nuclei in the ventral midbrain (VTA and SNpr) is distinct from the rest of midbrain, but the reason for this difference remained elusive. Here, we have analyzed the developmental origin of the VTA and SNpr GABAergic neurons by genetic fate mapping. We demonstrate that the majority of these GABAergic neurons originate outside the midbrain, from rhombomere 1, and move into the ventral midbrain only as postmitotic neuronal precursors. We further show that Gata2, Gata3 and Tal1 define a subpopulation of GABAergic precursors in ventral rhombomere 1. A failure in GABAergic neuron differentiation in this region correlates with loss of VTA and SNpr GABAergic neurons in Tal1 mutant mice. In contrast to midbrain, GABAergic neurons of the anterior SNpr in the diencephalon are not derived from the rhombomere 1. These results suggest unique migratory pathways for the precursors of important GABAergic neuron subpopulations, and provide the basis for understanding diversity within midbrain GABAergic neurons.


Subject(s)
Dopaminergic Neurons/physiology , GABAergic Neurons/physiology , Substantia Nigra/growth & development , Ventral Tegmental Area/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Cell Lineage , Cell Movement , Embryonic Development , Female , GATA2 Transcription Factor/analysis , GATA3 Transcription Factor/analysis , Mice , Proto-Oncogene Proteins/analysis , Substantia Nigra/cytology , T-Cell Acute Lymphocytic Leukemia Protein 1 , Ventral Tegmental Area/cytology
8.
Development ; 139(20): 3795-805, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22991444

ABSTRACT

Diverse mechanisms regulate development of GABAergic neurons in different regions of the central nervous system. We have addressed the roles of a proneural gene, Ascl1, and a postmitotic selector gene, Gata2, in the differentiation of GABAergic neuron subpopulations in three diencephalic prosomeres: prethalamus (P3), thalamus (P2) and pretectum (P1). Although the different proliferative progenitor populations of GABAergic neurons commonly express Ascl1, they have distinct requirements for it in promotion of cell-cycle exit and GABAergic neuron identity. Subsequently, Gata2 is activated as postmitotic GABAergic precursors are born. In P1, Gata2 regulates the neurotransmitter identity by promoting GABAergic and inhibiting glutamatergic neuron differentiation. Interestingly, Gata2 defines instead the subtype of GABAergic neurons in the rostral thalamus (pTh-R), which is a subpopulation of P2. Without Gata2, the GABAergic precursors born in the pTh-R fail to activate subtype-specific markers, but start to express genes typical of GABAergic precursors in the neighbouring P3 domain. Thus, our results demonstrate diverse mechanisms regulating differentiation of GABAergic neuron subpopulations and suggest a role for Gata2 as a selector gene of both GABAergic neuron neurotransmitter and prosomere subtype identities in the developing diencephalon. Our results demonstrate for the first time that neuronal identities between distinct prosomeres can still be transformed in postmitotic neuronal precursors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Diencephalon/embryology , GABAergic Neurons/metabolism , GATA2 Transcription Factor/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/embryology , Cell Differentiation , Diencephalon/cytology , GATA2 Transcription Factor/genetics , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , Neurogenesis , Thalamus/cytology , Thalamus/embryology , Transcriptional Activation
9.
Cell Mol Life Sci ; 71(8): 1395-415, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24196748

ABSTRACT

Neurons using gamma-aminobutyric acid (GABA) as their neurotransmitter are the main inhibitory neurons in the mature central nervous system (CNS) and show great variation in their form and function. GABAergic neurons are produced in all of the main domains of the CNS, where they develop from discrete regions of the neuroepithelium. Here, we review the gene expression and regulatory mechanisms controlling the main steps of GABAergic neuron development: early patterning of the proliferative neuroepithelium, production of postmitotic neural precursors, establishment of their identity and migration. By comparing the molecular regulation of these events across CNS, we broadly identify three regions utilizing distinct molecular toolkits for GABAergic fate determination: telencephalon-anterior diencephalon (DLX2 type), posterior diencephalon-midbrain (GATA2 type) and hindbrain-spinal cord (PTF1A and TAL1 types). Similarities and differences in the molecular regulatory mechanisms reveal the core determinants of a GABAergic neuron as well as provide insights into generation of the vast diversity of these neurons.


Subject(s)
Cell Differentiation/physiology , Central Nervous System/embryology , GABAergic Neurons/cytology , GABAergic Neurons/physiology , Gene Expression Regulation, Developmental/physiology , Models, Biological , Neurogenesis/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning/physiology , Cell Lineage/physiology , Cell Movement/physiology , Central Nervous System/cytology , GABAergic Neurons/classification , Gene Expression Regulation, Developmental/genetics , Humans , Proto-Oncogene Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/metabolism
10.
Nat Genet ; 36(9): 925-7, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15340424

ABSTRACT

The European Mouse Mutagenesis Consortium is the European initiative contributing to the international effort on functional annotation of the mouse genome. Its objectives are to establish and integrate mutagenesis platforms, gene expression resources, phenotyping units, storage and distribution centers and bioinformatics resources. The combined efforts will accelerate our understanding of gene function and of human health and disease.


Subject(s)
Genetic Research , Genome , Mice/genetics , Mutagenesis , Animals , Computational Biology , Europe , Phenotype
11.
FASEB Bioadv ; 5(5): 183-198, 2023 May.
Article in English | MEDLINE | ID: mdl-37151849

ABSTRACT

Snd1 is an evolutionarily conserved RNA-binding protein implicated in several regulatory processes in gene expression including activation of transcription, mRNA splicing, and microRNA decay. Here, we have investigated the outcome of Snd1 gene deletion in the mouse. The knockout mice are viable showing no gross abnormalities apart from decreased fertility, organ and body size, and decreased number of myeloid cells concomitant with decreased expression of granule protein genes. Deletion of Snd1 affected the expression of relatively small number of genes in spleen and liver. However, mRNA expression changes in the knockout mouse liver showed high similarity to expression profile in adaptation to hypoxia. MicroRNA expression in liver showed upregulation of the hypoxia-induced microRNAs miR-96 and -182. Similar to Snd1 deletion, mimics of miR-96/182 enhanced hypoxia-responsive reporter activity. To further elucidate the function of SND1, BioID biotin proximity ligation assay was performed in HEK-293T cells to identify interacting proteins. Over 50% of the identified interactors were RNA-binding proteins, including stress granule proteins. Taken together, our results show that in normal growth conditions, Snd1 is not a critical factor for mRNA transcription in the mouse, and describe a function for Snd1 in hypoxia adaptation through negatively regulating hypoxia-related miRNAs and hypoxia-induced transcription consistent with a role as stress response regulator.

12.
Dev Biol ; 349(2): 270-82, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21074523

ABSTRACT

For the correct development of the central nervous system, the balance between self-renewing and differentiating divisions of the neuronal progenitors must be tightly regulated. To maintain their self-renewing identity, the progenitors need to retain both apical and basal interfaces. However, the identities of fate-determining signals which cells receive via these connections, and the exact mechanism of their action, are poorly understood. The conditional inactivation of Fibroblast growth factor (FGF) receptors 1 and 2 in the embryonic mouse midbrain-hindbrain area results in premature neuronal differentiation. Here, we aim to elucidate the connection between FGF signaling and neuronal progenitor maintenance. Our results reveal that the loss of FGF signaling leads to downregulation of Hes1 and upregulation of Ngn2, Dll1, and p57 in the ventricular zone (VZ) cells, and that this increased neurogenesis occurs cell-autonomously. Yet the cell cycle progression, apico-basal-polarity, cell-cell connections, and the positioning of mitotic spindle in the mutant VZ appear unaltered. Interestingly, FGF8-protein is highly concentrated in the basal lamina. Thus, FGFs may act through basal processes of neuronal progenitors to maintain their progenitor status. Indeed, midbrain neuronal progenitors deprived in vitro of FGFs switched from symmetrical proliferative towards symmetrical neurogenic divisions. We suggest that FGF signaling in the midbrain VZ is cell-autonomously required for the maintenance of symmetrical proliferative divisions via Hes1-mediated repression of neurogenic genes.


Subject(s)
Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Mesencephalon/embryology , Neural Stem Cells/physiology , Neurogenesis/physiology , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine , Cell Proliferation , Crosses, Genetic , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Neurogenesis/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Transcription Factor HES-1
13.
Dev Biol ; 357(1): 259-68, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21736876

ABSTRACT

ß-catenin has well-established functions in cell growth and differentiation as part of the Wnt signaling pathway and in regulation of cellular adhesion with E-cadherin. Here we studied its significance in midbrain development by temporally controlled deletion of ß-catenin allowing simultaneous analysis of complete (ß-cat-null) and partial (ß-cat-low) loss-of-function phenotypes in progenitor cells. ß-cat-null cells did not contain centrosomes or a microtubule network and were unpolarized forming delaminated bulges. ß-cat-low cells displayed defects in the orientation of the mitotic spindle, increased asymmetric cell divisions and premature differentiation in absence of alterations in polarity or adhesion. The spindle defect was associated with decreased centrosomal S33/S34/T41 phosphorylated ß-catenin (p-ß-cat) and centrosomal and microtubule defects. Interestingly, neural progenitor cells in mice expressing only unphosphorylatable ß-catenin share several phenotypes with ß-catenin loss-of-function mice with defects in microtubules and polarity. The results demonstrate a novel function for p-ß-cat in maintaining neuroepithelial integrity and suggest that centrosomal p-ß-cat is required to maintain symmetric cleavages and polarity in neural progenitors.


Subject(s)
Centrosome/metabolism , Mesencephalon/embryology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , beta Catenin/metabolism , Animals , Cell Polarity/physiology , Dogs , Embryo, Mammalian/metabolism , Female , Mesencephalon/metabolism , Mice , Mice, Inbred Strains , Neural Stem Cells/cytology , Neurons/cytology , Phosphorylation , beta Catenin/analysis
14.
Dev Dyn ; 240(8): 1989-2001, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21761482

ABSTRACT

Classical cadherins are important cell adhesion molecules specifying and separating brain nuclei and developmental compartments. Cadherin-22 (Cdh22) belongs to type II subfamily of classical cadherins, and is expressed at the midbrain-hindbrain boundary during early embryogenesis. In Fgfr1 mutant mouse embryos, which have a disturbed midbrain-hindbrain border, Cdh22 is down-regulated. Here, we studied expression of Cdh22 in developing mouse brain in more detail and compared it to expression of related family members. This revealed both complementary and overlapping patterns of Cdh22, Cdh11, Cdh8, and Cdh6 expression in distinct regions of the forebrain and midbrain. We used a mutated allele of Cdh22 to study its function in brain development. Loss of Cdh22 caused reduced postnatal viability. Despite strong Cdh22 expression in the developing brain, we did not observe defects in compartmentalization or abnormalities in the midbrain and forebrain nuclei in Cdh22 mutants. This may be explained by functional redundancy between type II cadherins.


Subject(s)
Brain/anatomy & histology , Brain/embryology , Brain/metabolism , Cadherins/metabolism , Gene Expression Regulation, Developmental , Animals , Cadherins/genetics , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Extremities/anatomy & histology , Extremities/embryology , Genotype , Male , Mice , Morphogenesis/physiology , Mutation , Survival Rate , Testis/anatomy & histology , Testis/embryology , Testis/metabolism
15.
Front Neurosci ; 16: 976209, 2022.
Article in English | MEDLINE | ID: mdl-36148148

ABSTRACT

The Substantia Nigra pars reticulata (SNpr) is the major information output site of the basal ganglia network and instrumental for the activation and adjustment of movement, regulation of the behavioral state and response to reward. Due to both overlapping and unique input and output connections, the SNpr might also have signal integration capacity and contribute to action selection. How the SNpr regulates these multiple functions remains incompletely understood. The SNpr is located in the ventral midbrain and is composed primarily of inhibitory GABAergic projection neurons that are heterogeneous in their properties. In addition, the SNpr contains smaller populations of other neurons, including glutamatergic neurons. Here, we discuss regionalization of the SNpr, in particular the division of the SNpr neurons to anterior (aSNpr) and posterior (pSNpr) subtypes, which display differences in many of their features. We hypothesize that unique developmental and molecular characteristics of the SNpr neuron subtypes correlate with both region-specific connections and notable functional specializations of the SNpr. Variation in both the genetic control of the SNpr neuron development as well as signals regulating cell migration and axon guidance may contribute to the functional diversity of the SNpr neurons. Therefore, insights into the various aspects of differentiation of the SNpr neurons can increase our understanding of fundamental brain functions and their defects in neurological and psychiatric disorders, including movement and mood disorders, as well as epilepsy.

16.
Dev Biol ; 343(1-2): 63-70, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20417196

ABSTRACT

Midbrain GABAergic neurons regulate multiple aspects of behavior and play important roles in psychiatric and neurological disease. These neurons constitute several anatomical and functional subpopulations, but their molecular heterogeneity and developmental regulatory mechanisms are poorly understood. Here we have studied the involvement of the proneural gene Ascl1 in the development of the midbrain GABAergic neurons. Analysis of Ascl1 mutant mice demonstrated highly region-specific requirements for Ascl1 for development of different GABAergic neuron subpopulations. Ascl1 is dispensable for the development of the ventral-most midbrain GABAergic neurons associated with dopaminergic nuclei substantia nigra pars reticulata (SNpr) and ventral tegmental area (VTA) GABAergic neurons. In the ventrolateral midbrain, loss of Ascl1 results in markedly delayed neurogenesis in the midbrain domains m3-m5. Within this region, Ascl1 has a unique role in m4, where it also regulates glutamatergic neurogenesis. Our results suggest that the m3-m5 midbrain neuroepithelium gives rise to the GABAergic neuron groups located in the midbrain reticular formation and ventrolateral periaqueductal gray. In contrast to m3-m5, Ascl1 is absolutely required in the dorsal midbrain domains m1-m2, for generation of the GABAergic neurons populating the superior and inferior colliculi as well as dorsal periaqueductal gray. These studies demonstrate different molecular regulatory mechanisms for the distinct midbrain GABAergic neuron subpopulations. Also, our results have implications on understanding the origins of the various midbrain GABAergic neuron groups in the embryonic neuroepithelium.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Mesencephalon/metabolism , Neurons/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryo, Mammalian/metabolism , Female , Male , Mesencephalon/embryology , Mice , Mutation , RNA, Messenger/metabolism
17.
Gastroenterology ; 139(4): 1385-96, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20603121

ABSTRACT

BACKGROUND & AIMS: Fibroblast growth factor receptor 4 (FGFR4) controls bile acid metabolism and protects the liver from fibrosis, but the roles of FGFR1 and FGFR2 in the adult liver are largely unknown. We investigated the functions and mechanisms of action of these receptors in liver homeostasis, regeneration, and fibrosis. METHODS: We generated mice with hepatocytes that lack FGFR1 and FGFR2 and subjected them to acute and chronic carbon tetrachloride-induced liver injury and partial hepatectomy; mice were also injected with FGF7. We performed histology, histomorphometry, real-time reverse transcription polymerase chain reaction, and immunoblot analyses. RESULTS: In hepatocytes, loss of FGFR1 and FGFR2 eliminated responsiveness to FGF7 and related FGF family members but did not affect toxin-induced liver injury and fibrosis. However, mortality after partial hepatectomy increased because of severe hepatocyte necrosis. These effects appeared to be mediated by a failure of hepatocytes to induce the expression of the transcriptional regulators Dbp and Tef upon liver surgery; this affected expression of their target genes, which encode detoxifying cytochrome P450 enzymes. We found that Dbp and Tef expression was directly controlled by FGFR signaling in hepatocytes. As a consequence of the reduced expression of genes that control detoxification, the liver tissue that remained after partial hepatectomy failed to efficiently metabolize endogenous compounds and the drugs applied for anesthesia/analgesia. CONCLUSIONS: We identified a new, cytoprotective effect of FGFR1 and FGFR2 in the regenerating liver and suggest the use of recombinant FGF7 to increase survival of patients after surgical resection of large amounts of liver tissue.


Subject(s)
Inactivation, Metabolic , Liver Regeneration , Liver/drug effects , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Fibroblast Growth Factor, Type 2/physiology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Proliferation , Cells, Cultured , Cytoprotection , DNA-Binding Proteins/genetics , Hepatectomy , Hepatocytes/physiology , Liver/metabolism , Male , Mice , Transcription Factors/genetics
18.
Transl Psychiatry ; 11(1): 538, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663781

ABSTRACT

Early life stress (ELS) is a well-characterized risk factor for mood and anxiety disorders. GABAergic microcircuits in the amygdala are critically implicated in anxiety; however, whether their function is altered after ELS is not known. Here we identify a novel mechanism by which kainate receptors (KARs) modulate feedforward inhibition in the lateral amygdala (LA) and show that this mechanism is downregulated after ELS induced by maternal separation (MS). Specifically, we show that in control rats but not after MS, endogenous activity of GluK1 subunit containing KARs disinhibit LA principal neurons during activation of cortical afferents. GluK1 antagonism attenuated excitability of parvalbumin (PV)-expressing interneurons, resulting in loss of PV-dependent inhibitory control and an increase in firing of somatostatin-expressing interneurons. Inactivation of Grik1 expression locally in the adult amygdala reduced ongoing GABAergic transmission and was sufficient to produce a mild anxiety-like behavioral phenotype. Interestingly, MS and GluK1-dependent phenotypes showed similar gender specificity, being detectable in male but not female rodents. Our data identify a novel KAR-dependent mechanism for cell-type and projection-specific functional modulation of the LA GABAergic microcircuit and suggest that the loss of GluK1 KAR function contributes to anxiogenesis after ELS.


Subject(s)
Anxiety , Receptors, Kainic Acid , Stress, Psychological , Animals , Male , Rats , Amygdala/metabolism , Down-Regulation , Interneurons/metabolism , Maternal Deprivation , Receptors, Kainic Acid/metabolism
19.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-33983905

ABSTRACT

Synovial sarcoma is an aggressive malignancy with no effective treatments for patients with metastasis. The synovial sarcoma fusion SS18-SSX, which recruits the SWI/SNF-BAF chromatin remodeling and polycomb repressive complexes, results in epigenetic activation of FGF receptor (FGFR) signaling. In genetic FGFR-knockout models, culture, and xenograft synovial sarcoma models treated with the FGFR inhibitor BGJ398, we show that FGFR1, FGFR2, and FGFR3 were crucial for tumor growth. Transcriptome analyses of BGJ398-treated cells and histological and expression analyses of mouse and human synovial sarcoma tumors revealed prevalent expression of two ETS factors and FGFR targets, ETV4 and ETV5. We further demonstrate that ETV4 and ETV5 acted as drivers of synovial sarcoma growth, most likely through control of the cell cycle. Upon ETV4 and ETV5 knockdown, we observed a striking upregulation of DUX4 and its transcriptional targets that activate the zygotic genome and drive the atrophy program in facioscapulohumeral dystrophy patients. In addition to demonstrating the importance of inhibiting all three FGFRs, the current findings reveal potential nodes of attack for the cancer with the discovery of ETV4 and ETV5 as appropriate biomarkers and molecular targets, and activation of the embryonic DUX4 pathway as a promising approach to block synovial sarcoma tumors.


Subject(s)
Proto-Oncogene Proteins c-ets/metabolism , Sarcoma, Synovial/metabolism , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenylurea Compounds/pharmacology , Proto-Oncogene Proteins c-ets/genetics , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/deficiency , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Sarcoma, Synovial/genetics , Sarcoma, Synovial/pathology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Dev Cell ; 8(1): 85-95, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15621532

ABSTRACT

The epicardium regulates growth and survival of the underlying myocardium. This activity depends on intrinsic retinoic acid (RA) and erythropoietin signals. However, these signals do not act directly on the myocardium and instead are proposed to regulate the production of an unidentified soluble epicardial derived mitogen. Here, we show that Fgf9, Fgf16, and Fgf20 are expressed in the endocardium and epicardium and that RA can induce epicardial expression of Fgf9. Using knockout mice and an embryonic heart organ culture system, we show that endocardial and epicardial derived FGF signals regulate myocardial proliferation during midgestation heart development. We further show that this FGF signal is received by both FGF receptors 1 and 2 acting redundantly in the cardiomyoblast. In the absence of this signal, premature differentiation results in cellular hypertrophy and newborn mice develop a dilated cardiomyopathy. FGFs thus constitute all or part of the epicardial signal regulating myocardial growth and differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/radiation effects , Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental/physiology , Myocardium/metabolism , Signal Transduction/physiology , Age Factors , Animals , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Embryo, Mammalian , Fibroblast Growth Factor 9 , Fibroblast Growth Factors/classification , Fibroblast Growth Factors/deficiency , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/drug effects , Heart/embryology , Immunohistochemistry/methods , In Situ Hybridization/methods , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Myocardium/ultrastructure , Organ Culture Techniques/methods , RNA, Messenger/biosynthesis , Receptors, Fibroblast Growth Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL