Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 11(3): e0151862, 2016.
Article in English | MEDLINE | ID: mdl-26986975

ABSTRACT

The P2X7 receptor is a member of the P2X family of ligand-gated ion channels. A single-nucleotide polymorphism leading to a glutamine (Gln) by arginine (Arg) substitution at codon 460 of the purinergic P2X7 receptor (P2X7R) has been associated with mood disorders. No change in function (loss or gain) has been described for this SNP so far. Here we show that although the P2X7R-Gln460Arg variant per se is not compromised in its function, co-expression of wild-type P2X7R with P2X7R-Gln460Arg impairs receptor function with respect to calcium influx, channel currents and intracellular signaling in vitro. Moreover, co-immunoprecipitation and FRET studies show that the P2X7R-Gln460Arg variant physically interacts with P2X7R-WT. Specific silencing of either the normal or polymorphic variant rescues the heterozygous loss of function phenotype and restores normal function. The described loss of function due to co-expression, unique for mutations in the P2RX7 gene so far, explains the mechanism by which the P2X7R-Gln460Arg variant affects the normal function of the channel and may represent a mechanism of action for other mutations.


Subject(s)
Polymorphism, Single Nucleotide/genetics , Receptors, Purinergic P2X7/physiology , Blotting, Western , Calcium/metabolism , Calcium/physiology , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Immunoprecipitation , Patch-Clamp Techniques , Polymorphism, Single Nucleotide/physiology , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/metabolism , Signal Transduction/physiology
2.
Mol Cell Endocrinol ; 362(1-2): 29-38, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22659651

ABSTRACT

As a key regulator of the neuroendocrine stress axis and as a neuromodulator in the brain, the neuropeptide corticotropin-releasing hormone (CRH) plays an important role in various diseases of the central nervous system. Its cognate receptor CRH receptor type 1 (CRHR1) is a potential novel target for the therapeutic intervention in major depressive disorder. Therefore, a more precise understanding of involved intracellular signaling mechanisms is essential. The objective of this project was to identify specific target genes of CRHR1-mediated signaling pathways in the corticotrope cell line AtT-20 and in the neuronal cell line HN9 using microarray technology and qRT-PCR, respectively. In addition, we assessed the capacity of validated target genes to directly impact on CRHR1-dependent signaling using reporter assays. Thereby, we identified a set of CRHR1 downstream targets with diverging and cell type-specific roles which strengthen the role of CRH and CRHR1 as dynamic modulators of a variety of signal transduction mechanisms and cellular processes.


Subject(s)
Corticotrophs/metabolism , Corticotropin-Releasing Hormone/physiology , Gene Expression Regulation , Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Cell Line, Tumor , Genes, Reporter , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Mice , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Response Elements , Transcription, Genetic , Transcriptional Activation , Transcriptome
3.
PLoS One ; 6(8): e23604, 2011.
Article in English | MEDLINE | ID: mdl-21897848

ABSTRACT

Behavioral endophenotypes are determined by a multitude of counteracting but precisely balanced molecular and physiological mechanisms. In this study, we aim to identify potential novel molecular targets that contribute to the multigenic trait "anxiety". We used microarrays to investigate the gene expression profiles of different brain regions within the limbic system of mice which were selectively bred for either high (HAB) or low (LAB) anxiety-related behavior, and also show signs of comorbid depression-like behavior. We identified and confirmed sex-independent differences in the basal expression of 13 candidate genes, using tissue from the entire brain, including coronin 7 (Coro7), cathepsin B (Ctsb), muscleblind-like 1 (Mbnl1), metallothionein 1 (Mt1), solute carrier family 25 member 17 (Slc25a17), tribbles homolog 2 (Trib2), zinc finger protein 672 (Zfp672), syntaxin 3 (Stx3), ATP-binding cassette, sub-family A member 2 (Abca2), ectonucleotide pyrophosphatase/phosphodiesterase 5 (Enpp5), high mobility group nucleosomal binding domain 3 (Hmgn3) and pyruvate dehydrogenase beta (Pdhb). Additionally, we confirmed brain region-specific differences in the expression of synaptotagmin 4 (Syt4).Our identification of about 90 polymorphisms in Ctsb suggested that this gene might play a critical role in shaping our mouse model's behavioral endophenotypes. Indeed, the assessment of anxiety-related and depression-like behaviors of Ctsb knock-out mice revealed an increase in depression-like behavior in females. Altogether, our results suggest that Ctsb has significant effects on emotionality, irrespective of the tested mouse strain, making it a promising target for future pharmacotherapy.


Subject(s)
Anxiety/enzymology , Anxiety/genetics , Cathepsin B/genetics , Gene Expression Profiling , Animals , Behavior, Animal , Brain/metabolism , Cathepsin B/deficiency , Endophenotypes , Female , Gene Knockout Techniques , In Situ Hybridization , Male , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Sequence Analysis, DNA
4.
BMC Genomics ; 11: 546, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20932279

ABSTRACT

BACKGROUND: The pivotal role of stress in the precipitation of psychiatric diseases such as depression is generally accepted. This study aims at the identification of genes that are directly or indirectly responding to stress. Inbred mouse strains that had been evidenced to differ in their stress response as well as in their response to antidepressant treatment were chosen for RNA profiling after stress exposure. Gene expression and regulation was determined by microarray analyses and further evaluated by bioinformatics tools including pathway and cluster analyses. RESULTS: Forced swimming as acute stressor was applied to C57BL/6J and DBA/2J mice and resulted in sets of regulated genes in the paraventricular nucleus of the hypothalamus (PVN), 4 h or 8 h after stress. Although the expression changes between the mouse strains were quite different, they unfolded in phases over time in both strains. Our search for connections between the regulated genes resulted in potential novel signalling pathways in stress. In particular, Guanine nucleotide binding protein, alpha inhibiting 2 (GNAi2) and amyloid ß (A4) precursor protein (APP) were detected as stress-regulated genes, and together with other genes, seem to be integrated into stress-responsive pathways and gene networks in the PVN. CONCLUSIONS: This search for stress-regulated genes in the PVN revealed its impact on interesting genes (GNAi2 and APP) and a novel gene network. In particular the expression of APP in the PVN that is governing stress hormone balance, is of great interest. The reported neuroprotective role of this molecule in the CNS supports the idea that a short acute stress can elicit positive adaptational effects in the brain.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Gene Expression Profiling , Gene Regulatory Networks/genetics , Paraventricular Hypothalamic Nucleus/metabolism , Stress, Physiological/genetics , Adrenocorticotropic Hormone/blood , Amyloid beta-Protein Precursor/metabolism , Animals , Cluster Analysis , Down-Regulation/genetics , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Reproducibility of Results , Signal Transduction/genetics , Species Specificity , Swimming/physiology , Up-Regulation/genetics
5.
Front Mol Neurosci ; 2: 1, 2009.
Article in English | MEDLINE | ID: mdl-19506703

ABSTRACT

The postnatal development of the mouse is characterized by a stress hypo-responsive period (SHRP), where basal corticosterone levels are low and responsiveness to mild stressors is reduced. Maternal separation is able to disrupt the SHRP and is widely used to model early trauma. In this study we aimed at identifying of brain systems involved in acute and possible long-term effects of maternal separation. We conducted a microarray-based gene expression analysis in the hypothalamic paraventricular nucleus after maternal separation, which revealed 52 differentially regulated genes compared to undisturbed controls, among them are 37 up-regulated and 15 down-regulated genes. One of the prominently up-regulated genes, angiotensinogen, was validated using in-situ hybridization. Angiotensinogen is the precursor of angiotensin II, the main effector of the brain renin-angiotensin system (RAS), which is known to be involved in stress system modulation in adult animals. Using the selective angiotensin type I receptor [AT(1)] antagonist candesartan we found strong effects on CRH and GR mRNA expression in the brain and ACTH release following maternal separation. AT(1) receptor blockade appears to enhance central effects of maternal separation in the neonate, suggesting a suppressing function of brain RAS during the SHRP. Taken together, our results illustrate the molecular adaptations that occur in the paraventricular nucleus following maternal separation and contribute to identifying signaling cascades that control stress system activity in the neonate.

6.
PLoS One ; 4(4): e5129, 2009.
Article in English | MEDLINE | ID: mdl-19357765

ABSTRACT

BACKGROUND: To investigate neurobiological correlates of trait anxiety, CD1 mice were selectively bred for extremes in anxiety-related behavior, with high (HAB) and low (LAB) anxiety-related behavior mice additionally differing in behavioral tests reflecting depression-like behavior. METHODOLOGY/ PRINCIPAL FINDINGS: In this study, microarray analysis, in situ hybridization, quantitative real-time PCR and immunohistochemistry revealed decreased expression of the vasopressin gene (Avp) in the hypothalamic paraventricular (PVN) and supraoptic (SON) nuclei of adult LAB mice compared to HAB, NAB (normal anxiety-related behavior) and HABxLAB F1 intercross controls, without detecting differences in receptor expression or density. By sequencing the regions 2.5 kbp up- and downstream of the Avp gene locus, we could identify several polymorphic loci, differing between the HAB and LAB lines. In the gene promoter, a deletion of twelve bp Delta(-2180-2191) is particularly likely to contribute to the reduced Avp expression detected in LAB animals under basal conditions. Indeed, allele-specific transcription analysis of F1 animals revealed a hypomorphic LAB-specific Avp allele with a reduced transcription rate by 75% compared to the HAB-specific allele, thus explaining line-specific Avp expression profiles and phenotypic features. Accordingly, intra-PVN Avp mRNA levels were found to correlate with anxiety-related and depression-like behaviors. In addition to this correlative evidence, a significant, though moderate, genotype/phenotype association was demonstrated in 258 male mice of a freely-segregating F2 panel, suggesting a causal contribution of the Avp promoter deletion to anxiety-related behavior. DISCUSSION: Thus, the identification of polymorphisms in the Avp gene promoter explains gene expression differences in association with the observed phenotype, thus further strengthening the concept of the critical involvement of centrally released AVP in trait anxiety.


Subject(s)
Alleles , Anxiety/genetics , Arginine Vasopressin/genetics , Behavior, Animal/physiology , Animals , Anxiety/physiopathology , Arginine Vasopressin/metabolism , Depression/genetics , Depression/physiopathology , Female , Gene Expression Profiling , Humans , Male , Mice , Motor Activity/physiology , Neuropsychological Tests , Oligonucleotide Array Sequence Analysis , Oxytocin/genetics , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Supraoptic Nucleus/metabolism
7.
Psychopharmacology (Berl) ; 200(4): 557-72, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18629477

ABSTRACT

INTRODUCTION: Monoamine-based antidepressants inhibit neurotransmitter reuptake within short time. However, it commonly takes several weeks until clinical symptoms start to resolve--indicating the involvement of effects distant from reuptake inhibition. OBJECTIVE: To unravel other mechanisms involved in drug action, a "reverse" pharmacological approach was applied to determine antidepressant-induced alterations of hippocampal gene expression. MATERIALS AND METHODS: The behavioral response to long-term paroxetine administration of male DBA/2Ola mice was assessed by the forced swim test (FST), the modified hole board (mHB), and the dark/light box. Hippocampi of test-naive mice were dissected, and changes in gene expression by paroxetine treatment were investigated by means of microarray technology. RESULTS AND DISCUSSION: Robust effects of paroxetine on passive stress-coping behavior in the FST were observed. Furthermore, anxiolytic properties of long-term antidepressant treatment could be identified in DBA mice in both, the mHB and dark/light box. Analysis of microarray results revealed a list of 60 genes differentially regulated by chronic paroxetine treatment. Preproenkephalin 1 and inhibin beta-A showed the highest level of transcriptional change. Furthermore, a number of candidates involved in neuroplasticity/neurogenesis emerged (e.g., Bdnf, Gfap, Vim, Sox11, Egr1, Stat3). Seven selected candidates were confirmed by in situ hybridization. Additional immunofluorescence colocalization studies of GFAP and vimentin showed more positive cells to be detected in long-term paroxetine-treated DBA mice. CONCLUSION: Candidate genes identified in the current study using a mouse strain validated for its responsiveness to long-term paroxetine treatment add, in our opinion, to unraveling the mechanism of action of paroxetine as a representative for SSRIs.


Subject(s)
Behavior, Animal/drug effects , Gene Expression Regulation/drug effects , Paroxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Antidepressive Agents, Second-Generation/pharmacology , Darkness , Depression/drug therapy , Depression/physiopathology , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Light , Male , Mice , Mice, Inbred DBA , Oligonucleotide Array Sequence Analysis/methods , Paroxetine/administration & dosage , Selective Serotonin Reuptake Inhibitors/administration & dosage , Swimming
8.
PLoS One ; 2(10): e1064, 2007 Oct 24.
Article in English | MEDLINE | ID: mdl-17957248

ABSTRACT

BACKGROUND: Phototrophy of the extremely halophilic archaeon Halobacterium salinarum was explored for decades. The research was mainly focused on the expression of bacteriorhodopsin and its functional properties. In contrast, less is known about genome wide transcriptional changes and their impact on the physiological adaptation to phototrophy. The tool of choice to record transcriptional profiles is the DNA microarray technique. However, the technique is still rarely used for transcriptome analysis in archaea. METHODOLOGY/PRINCIPAL FINDINGS: We developed a whole-genome DNA microarray based on our sequence data of the Hbt. salinarum strain R1 genome. The potential of our tool is exemplified by the comparison of cells growing under aerobic and phototrophic conditions, respectively. We processed the raw fluorescence data by several stringent filtering steps and a subsequent MAANOVA analysis. The study revealed a lot of transcriptional differences between the two cell states. We found that the transcriptional changes were relatively weak, though significant. Finally, the DNA microarray data were independently verified by a real-time PCR analysis. CONCLUSION/SIGNIFICANCE: This is the first DNA microarray analysis of Hbt. salinarum cells that were actually grown under phototrophic conditions. By comparing the transcriptomics data with current knowledge we could show that our DNA microarray tool is well applicable for transcriptome analysis in the extremely halophilic archaeon Hbt. salinarum. The reliability of our tool is based on both the high-quality array of DNA probes and the stringent data handling including MAANOVA analysis. Among the regulated genes more than 50% had unknown functions. This underlines the fact that haloarchaeal phototrophy is still far away from being completely understood. Hence, the data recorded in this study will be subject to future systems biology analysis.


Subject(s)
Gene Expression Regulation, Bacterial , Halobacterium salinarum/genetics , Halobacterium salinarum/metabolism , Oligonucleotide Array Sequence Analysis , Archaeal Proteins/genetics , Biochemistry/methods , DNA Primers/chemistry , DNA, Complementary/metabolism , Genome, Bacterial , Light , Models, Statistical , Nucleic Acid Hybridization , RNA/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Fluorescence , Transcription, Genetic
9.
J Cereb Blood Flow Metab ; 27(8): 1476-95, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17293846

ABSTRACT

Corticotropin-releasing hormone receptor type 1 (CRH-R1)-deficient mice display reduced anxiety-like behavior, a chronic corticosterone deficit, and an impaired neuroendocrine stress response caused by disruption of the hypothalamic-pituitary-adrenocortical (HPA) axis. The molecular substrates and pathways of CRH/CRH-R1-dependent signaling mechanisms underlying the behavioral phenotype as well as the consequences of lifelong glucocorticoid deficit remain largely obscure. To dissect involved neuronal circuitries, we performed comparative expression profiling of brains of CRH-R1 mutant and wild-type mice using our custom made MPIP (Max Planck Institute of Psychiatry) 17k cDNA microarray. Microarray analysis yielded 107 genes showing altered expression levels when comparing CRH-R1 knockout mice with wild-type littermates. A significant proportion of differentially expressed genes was related to control of HPA and hypothalamic-pituitary-thyroid (HPT) axes reflecting not only the disturbance of the HPA axis in CRH-R1 mutant mice but also the interplay of both neuroendocrine systems. The spatial analysis of regulated genes revealed a prevalence for genes expressed in the cerebral microvasculature. This phenotype was confirmed by the successful cross-validation of regulated genes in CRH overexpressing mice. Analysis of the cerebral vasculature of CRH-R1 mutant and CRH overexpressing mice revealed alterations of functional rather than structural properties. A direct role of the CRH/CRH-R1 system was supported by demonstrating Crhr1 expression in the adult murine cerebral vasculature. In conclusion, these data suggest a novel, previously unknown role of the CRH/CRH-R1 system in modulating neurovascular gene expression and function.


Subject(s)
Brain/blood supply , Corticotropin-Releasing Hormone/metabolism , Gene Expression Regulation , Receptors, Corticotropin-Releasing Hormone/metabolism , Alkaline Phosphatase/metabolism , Animals , Behavior, Animal/physiology , Brain/metabolism , Cerebrovascular Circulation/physiology , Gene Expression Profiling , Glucocorticoids/metabolism , Hypothalamo-Hypophyseal System/physiology , In Situ Hybridization , Male , Mice , Mice, Knockout , Microcirculation , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pituitary-Adrenal System/physiology , Receptors, Corticotropin-Releasing Hormone/genetics , Reproducibility of Results , Signal Transduction/physiology
10.
Cancer Res ; 66(24): 11975-82, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178896

ABSTRACT

Zac is a C2H2 zinc finger protein, which regulates apoptosis and cell cycle arrest through DNA binding and transactivation. During tumorigenesis and in response to mitogenic activation, Zac gene expression is down-regulated in a methylation-sensitive manner. As yet, no target genes have been identified that could explain the potent antiproliferative function of Zac. Here, applying genome-wide expression analysis, we identify peroxisome proliferator-activated receptor gamma (PPARgamma) as a new bona fide Zac target gene, which is induced by direct Zac binding to the proximal PPARgamma1 promoter. We show that in human colon carcinoma cells, ZAC activates expression of PPARgamma target genes in a PPARgamma-dependent manner. Moreover, we show that treatment of pituitary tumor cells with octreotide, a somatostatin analogue, leads to Zac induction and subsequent Zac-dependent up-regulation of PPARgamma, which thereupon mediates part of the antiproliferative activity of Zac. Our work provides a first step toward elucidating a functional relationship between Zac and PPARgamma that could be relevant to the understanding of tumorigenesis and diabetes as well.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression Regulation , PPAR gamma/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Cell Line, Tumor , Cloning, Molecular , Colonic Neoplasms , Humans , Oligonucleotide Array Sequence Analysis , Pituitary Neoplasms , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
11.
Development ; 133(1): 89-98, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16339193

ABSTRACT

Midbrain neurons synthesizing the neurotransmitter dopamine play a central role in the modulation of different brain functions and are associated with major neurological and psychiatric disorders. Despite the importance of these cells, the molecular mechanisms controlling their development are still poorly understood. The secreted glycoprotein Wnt1 is expressed in close vicinity to developing midbrain dopaminergic neurons. Here, we show that Wnt1 regulates the genetic network, including Otx2 and Nkx2-2, that is required for the establishment of the midbrain dopaminergic progenitor domain during embryonic development. In addition, Wnt1 is required for the terminal differentiation of midbrain dopaminergic neurons at later stages of embryogenesis. These results identify Wnt1 as a key molecule in the development of midbrain dopaminergic neurons in vivo. They also suggest the Wnt1-controlled signaling pathway as a promising target for new therapeutic strategies in the treatment of Parkinson's disease.


Subject(s)
Cell Differentiation/physiology , Mesencephalon/embryology , Neurons/physiology , Signal Transduction/physiology , Stem Cells/physiology , Wnt1 Protein/metabolism , Animals , Homeobox Protein Nkx-2.2 , Immunohistochemistry , In Situ Hybridization , Mesencephalon/metabolism , Mice , Mice, Transgenic , Microspheres , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism
12.
J Neurosci ; 25(17): 4375-84, 2005 Apr 27.
Article in English | MEDLINE | ID: mdl-15858064

ABSTRACT

For >15 generations, CD1 mice have been selectively and bidirectionally bred for either high-anxiety-related behavior (HAB-M) or low-anxiety-related behavior (LAB-M) on the elevated plus-maze. Independent of gender, HAB-M were more anxious than LAB-M animals in a variety of additional tests, including those reflecting risk assessment behaviors and ultrasound vocalization, with unselected CD1 "normal" control (NAB-M) and cross-mated (CM-M) mice displaying intermediate behavioral scores in most cases. Furthermore, in both the forced-swim and tail-suspension tests, LAB-M animals showed lower scores of immobility than did HAB-M and NAB-M animals, indicative of a reduced depression-like behavior. Using proteomic and microarray analyses, glyoxalase-I was identified as a protein marker, which is consistently expressed to a higher extent in LAB-M than in HAB-M mice in several brain areas. The same phenotype-dependent difference was found in red blood cells with NAB-M and CM-M animals showing intermediate expression profiles of glyoxalase-I. Additional studies will examine whether glyoxalase-I has an impact beyond that of a biomarker to predict the genetic predisposition to anxiety- and depression-like behavior.


Subject(s)
Anxiety Disorders/enzymology , Lactoylglutathione Lyase/metabolism , Analysis of Variance , Animals , Animals, Newborn , Anti-Anxiety Agents/administration & dosage , Anxiety Disorders/diagnosis , Anxiety Disorders/drug therapy , Anxiety Disorders/genetics , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal , Biomarkers/metabolism , Blotting, Western/methods , Brain/drug effects , Brain/metabolism , Breeding/methods , Diazepam/administration & dosage , Disease Models, Animal , Electrophoresis, Gel, Two-Dimensional/methods , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Hindlimb Suspension/physiology , Lactoylglutathione Lyase/isolation & purification , Locomotion/genetics , Male , Mass Spectrometry/methods , Mice , Microarray Analysis/methods , Phenotype , Predictive Value of Tests , Proteomics/methods , Reaction Time/physiology , Reproducibility of Results , Sex Factors , Spatial Behavior/drug effects , Spatial Behavior/physiology , Statistics as Topic , Swimming , Time Factors , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
13.
Mol Cell Neurosci ; 26(1): 101-11, 2004 May.
Article in English | MEDLINE | ID: mdl-15121182

ABSTRACT

The secreted glycoprotein WNT1 is expressed in the caudal midbrain and is essential for proper development of the entire mid-/hindbrain region. To get better insights into Wnt1 function in the mid-/hindbrain region, we ectopically expressed Wnt1 under the control of the endogenous En1 promoter, thereby extending Wnt1 expression rostrally into the anterior midbrain and caudally into rhombomere 1. In these transgenic mice, the position of the mid-/hindbrain organizer is not altered and pattern formation is not changed. During midgestation, ectopic Wnt1 induced strong overproliferation of precursor cells only in the caudal midbrain in a gene dosage-dependent manner. Enhanced proliferation is at least in part mediated by shortening of the cell cycle length. In adults, Wnt1 exhibited a cell size promoting effect specifically on neurons. We suggest that Wnt1 acts as a regulator of proliferation of specific precursor populations in the developing mid-/hindbrain region and is only secondarily involved in maintenance of the mid-/hindbrain organizer.


Subject(s)
Mesencephalon/embryology , Neurons/metabolism , Proto-Oncogene Proteins/metabolism , Rhombencephalon/embryology , Stem Cells/metabolism , Animals , Body Patterning/genetics , Cell Differentiation/genetics , Cell Division/genetics , Female , Fetus , Gene Dosage , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/genetics , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Mice, Transgenic , Neurons/cytology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Rhombencephalon/cytology , Rhombencephalon/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stem Cells/cytology , Wnt Proteins , Wnt1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...