Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Cell Rep ; 37(2): 109802, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34644582

ABSTRACT

Tissue-clearing methods allow every cell in the mouse brain to be imaged without physical sectioning. However, the computational tools currently available for cell quantification in cleared tissue images have been limited to counting sparse cell populations in stereotypical mice. Here, we introduce NuMorph, a group of analysis tools to quantify all nuclei and nuclear markers within the mouse cortex after clearing and imaging by light-sheet microscopy. We apply NuMorph to investigate two distinct mouse models: a Topoisomerase 1 (Top1) model with severe neurodegenerative deficits and a Neurofibromin 1 (Nf1) model with a more subtle brain overgrowth phenotype. In each case, we identify differential effects of gene deletion on individual cell-type counts and distribution across cortical regions that manifest as alterations of gross brain morphology. These results underline the value of whole-brain imaging approaches, and the tools are widely applicable for studying brain structure phenotypes at cellular resolution.


Subject(s)
Cell Nucleus/pathology , Cerebral Cortex/pathology , Histocytological Preparation Techniques , Nerve Degeneration , Neuroglia/pathology , Neuroimaging , Neurons/pathology , Animals , Cell Nucleus/metabolism , Cerebral Cortex/metabolism , DNA Topoisomerases, Type I/deficiency , DNA Topoisomerases, Type I/genetics , Gene Deletion , Genes, Neurofibromatosis 1 , Image Processing, Computer-Assisted , Mice, Knockout , Neuroglia/metabolism , Neurons/metabolism , Phenotype , Support Vector Machine
2.
Sci Rep ; 11(1): 20293, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645873

ABSTRACT

Cryptochromes (CRY) are highly conserved signalling molecules that regulate circadian rhythms and are candidate radical pair based magnetoreceptors. Birds have at least four cryptochromes (CRY1a, CRY1b, CRY2, and CRY4), but few studies have interrogated their function. Here we investigate the expression, localisation and interactome of clCRY2 in the pigeon retina. We report that clCRY2 has two distinct transcript variants, clCRY2a, and a previously unreported splice isoform, clCRY2b which is larger in size. We show that clCRY2a mRNA is expressed in all retinal layers and clCRY2b is enriched in the inner and outer nuclear layer. To define the localisation and interaction network of clCRY2 we generated and validated a monoclonal antibody that detects both clCRY2 isoforms. Immunohistochemical studies revealed that clCRY2a/b is present in all retinal layers and is enriched in the outer limiting membrane and outer plexiform layer. Proteomic analysis showed clCRY2a/b interacts with typical circadian molecules (PER2, CLOCK, ARTNL), cell junction proteins (CTNNA1, CTNNA2) and components associated with the microtubule motor dynein (DYNC1LI2, DCTN1, DCTN2, DCTN3) within the retina. Collectively these data show that clCRY2 is a component of the avian circadian clock and unexpectedly associates with the microtubule cytoskeleton.


Subject(s)
Cryptochromes/metabolism , Microtubules/metabolism , Retina/metabolism , Alternative Splicing , Animals , Circadian Clocks , Circadian Rhythm/physiology , Cloning, Molecular , Columbidae/metabolism , Genetic Variation , Intercellular Junctions , Mass Spectrometry , Protein Isoforms , Proteomics/methods , Retina/pathology
3.
Cereb Cortex ; 31(6): 3064-3081, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33570093

ABSTRACT

Many developmental syndromes have been linked to genetic mutations that cause abnormal ERK/MAPK activity; however, the neuropathological effects of hyperactive signaling are not fully understood. Here, we examined whether hyperactivation of MEK1 modifies the development of GABAergic cortical interneurons (CINs), a heterogeneous population of inhibitory neurons necessary for cortical function. We show that GABAergic-neuron specific MEK1 hyperactivation in vivo leads to increased cleaved caspase-3 labeling in a subpopulation of immature neurons in the embryonic subpallial mantle zone. Adult mutants displayed a significant loss of parvalbumin (PV), but not somatostatin, expressing CINs and a reduction in perisomatic inhibitory synapses on excitatory neurons. Surviving mutant PV-CINs maintained a typical fast-spiking phenotype but showed signs of decreased intrinsic excitability that coincided with an increased risk of seizure-like phenotypes. In contrast to other mouse models of PV-CIN loss, we discovered a robust increase in the accumulation of perineuronal nets, an extracellular structure thought to restrict plasticity. Indeed, we found that mutants exhibited a significant impairment in the acquisition of behavioral response inhibition capacity. Overall, our data suggest PV-CIN development is particularly sensitive to hyperactive MEK1 signaling, which may underlie certain neurological deficits frequently observed in ERK/MAPK-linked syndromes.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , GABAergic Neurons/metabolism , Inhibition, Psychological , MAP Kinase Kinase 1/metabolism , Parvalbumins/metabolism , Animals , Cerebral Cortex/chemistry , Electroencephalography/methods , Embryonic Development/physiology , GABAergic Neurons/chemistry , Locomotion/physiology , MAP Kinase Kinase 1/analysis , Mice , Organ Culture Techniques , Parvalbumins/analysis , Signal Transduction/physiology
4.
Sci Adv ; 6(33): eabb9110, 2020 08.
Article in English | MEDLINE | ID: mdl-32851187

ABSTRACT

The biophysical and molecular mechanisms that enable animals to detect magnetic fields are unknown. It has been proposed that birds have a light-dependent magnetic compass that relies on the formation of radical pairs within cryptochrome molecules. Using spectroscopic methods, we show that pigeon cryptochrome clCRY4 is photoreduced efficiently and forms long-lived spin-correlated radical pairs via a tetrad of tryptophan residues. We report that clCRY4 is broadly and stably expressed within the retina but enriched at synapses in the outer plexiform layer in a repetitive manner. A proteomic survey for retinal-specific clCRY4 interactors identified molecules that are involved in receptor signaling, including glutamate receptor-interacting protein 2, which colocalizes with clCRY4. Our data support a model whereby clCRY4 acts as an ultraviolet-blue photoreceptor and/or a light-dependent magnetosensor by modulating glutamatergic synapses between horizontal cells and cones.

5.
PLoS Genet ; 15(4): e1008108, 2019 04.
Article in English | MEDLINE | ID: mdl-31017896

ABSTRACT

RASopathies are a family of related syndromes caused by mutations in regulators of the RAS/Extracellular Regulated Kinase 1/2 (ERK1/2) signaling cascade that often result in neurological deficits. RASopathy mutations in upstream regulatory components, such as NF1, PTPN11/SHP2, and RAS have been well-characterized, but mutation-specific differences in the pathogenesis of nervous system abnormalities remain poorly understood, especially those involving mutations downstream of RAS. Here, we assessed cellular and behavioral phenotypes in mice expressing a Raf1L613V gain-of-function mutation associated with the RASopathy, Noonan Syndrome. We report that Raf1L613V/wt mutants do not exhibit a significantly altered number of excitatory or inhibitory neurons in the cortex. However, we observed a significant increase in the number of specific glial subtypes in the forebrain. The density of GFAP+ astrocytes was significantly increased in the adult Raf1L613V/wt cortex and hippocampus relative to controls. OLIG2+ oligodendrocyte progenitor cells were also increased in number in mutant cortices, but we detected no significant change in myelination. Behavioral analyses revealed no significant changes in voluntary locomotor activity, anxiety-like behavior, or sociability. Surprisingly, Raf1L613V/wt mice performed better than controls in select aspects of the water radial-arm maze, Morris water maze, and cued fear conditioning tasks. Overall, these data show that increased astrocyte and oligodendrocyte progenitor cell (OPC) density in the cortex coincides with enhanced cognition in Raf1L613V/wt mutants and further highlight the distinct effects of RASopathy mutations on nervous system development and function.


Subject(s)
Cerebral Cortex/metabolism , Learning , Mutation , Neuroglia/metabolism , Noonan Syndrome/genetics , Noonan Syndrome/psychology , Proto-Oncogene Proteins c-raf/genetics , Animals , Biomarkers , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , MAP Kinase Signaling System , Maze Learning , Memory , Mice , Mice, Transgenic , Neurons/metabolism , Noonan Syndrome/metabolism , Oligodendroglia/metabolism , Proto-Oncogene Proteins c-raf/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L784-L797, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30724100

ABSTRACT

A defining characteristic of pulmonary hypertension (PH) is the extensive remodeling of pulmonary arteries (PAs), which results in progressive increases in vascular resistance and stiffness and eventual failure of the right ventricle. There is no cure for PH and identification of novel molecular mechanisms that underlie increased proliferation, reduced apoptosis, and excessive extracellular matrix production in pulmonary artery smooth muscle cells (PASMCs) is a vital objective. Galectin-3 (Gal-3) is a chimeric lectin and potent driver of many aspects of fibrosis, but its role in regulating PASMC behavior in PH remains poorly understood. Herein, we evaluated the importance of increased Gal-3 expression and signaling on PA vascular remodeling and cardiopulmonary function in experimental models of PH. Gal-3 expression was quantified by qRT-PCR, immunoblotting, and immunofluorescence imaging, and its functional role was assessed by specific Gal-3 inhibitors and CRISPR/Cas9-mediated knockout of Gal-3 in the rat. In rat models of PH, we observed increased Gal-3 expression in PASMCs, which stimulated migration and resistance to apoptosis, whereas silencing or genetic deletion reduced cellular migration and PA fibrosis and increased apoptosis. Gal-3 inhibitors attenuated and reversed PA remodeling and fibrosis, as well as hemodynamic indices in monocrotaline (MCT)-treated rats in vivo. These results were supported by genetic deletion of Gal-3 in both MCT and Sugen Hypoxia rat models. In conclusion, our results suggest that elevated Gal-3 levels contribute to inappropriate PA remodeling in PH by enhancing multiple profibrotic mechanisms. Therapeutic strategies targeting Gal-3 may be of benefit in the treatment of PH.


Subject(s)
Apoptosis , Cell Proliferation , Galectin 3/biosynthesis , Gene Expression Regulation , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Fibrosis/metabolism , Animals , Blood Proteins , Disease Models, Animal , Galectin 3/genetics , Galectins , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley
7.
J Neurosci ; 37(34): 8102-8115, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28733355

ABSTRACT

The ERK/MAPK intracellular signaling pathway is hypothesized to be a key regulator of striatal activity via modulation of synaptic plasticity and gene transcription. However, prior investigations into striatal ERK/MAPK functions have yielded conflicting results. Further, these studies have not delineated the cell-type-specific roles of ERK/MAPK signaling due to the reliance on globally administered pharmacological ERK/MAPK inhibitors and the use of genetic models that only partially reduce total ERK/MAPK activity. Here, we generated mouse models in which ERK/MAPK signaling was completely abolished in each of the two distinct classes of medium spiny neurons (MSNs). ERK/MAPK deletion in D1R-MSNs (direct pathway) resulted in decreased locomotor behavior, reduced weight gain, and early postnatal lethality. In contrast, loss of ERK/MAPK signaling in D2R-MSNs (indirect pathway) resulted in a profound hyperlocomotor phenotype. ERK/MAPK-deficient D2R-MSNs exhibited a significant reduction in dendritic spine density, markedly suppressed electrical excitability, and suppression of activity-associated gene expression even after pharmacological stimulation. Our results demonstrate the importance of ERK/MAPK signaling in governing the motor functions of the striatal direct and indirect pathways. Our data further show a critical role for ERK in maintaining the excitability and plasticity of D2R-MSNs.SIGNIFICANCE STATEMENT Alterations in ERK/MAPK activity are associated with drug abuse, as well as neuropsychiatric and movement disorders. However, genetic evidence defining the functions of ERK/MAPK signaling in striatum-related neurophysiology and behavior is lacking. We show that loss of ERK/MAPK signaling leads to pathway-specific alterations in motor function, reduced neuronal excitability, and the inability of medium spiny neurons to regulate activity-induced gene expression. Our results underscore the potential importance of the ERK/MAPK pathway in human movement disorders.


Subject(s)
Corpus Striatum/physiology , Locomotion/physiology , MAP Kinase Signaling System/physiology , Movement/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation
8.
Cell Rep ; 18(13): 3167-3177, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28355568

ABSTRACT

During development of the vertebrate CNS, the basic helix-loop-helix (bHLH) transcription factor Olig2 sustains replication competence of progenitor cells that give rise to neurons and oligodendrocytes. A pathological counterpart of this developmental function is seen in human glioma, wherein Olig2 is required for maintenance of stem-like cells that drive tumor growth. The mitogenic/gliomagenic functions of Olig2 are regulated by phosphorylation of a triple serine motif (S10, S13, and S14) in the amino terminus. Here, we identify a set of three serine/threonine protein kinases (glycogen synthase kinase 3α/ß [GSK3α/ß], casein kinase 2 [CK2], and cyclin-dependent kinases 1/2 [CDK1/2]) that are, collectively, both necessary and sufficient to phosphorylate the triple serine motif. We show that phosphorylation of the motif itself serves as a template to prime phosphorylation of additional serines and creates a highly charged "acid blob" in the amino terminus of Olig2. Finally, we show that small molecule inhibitors of this forward-feeding phosphorylation cascade have potential as glioma therapeutics.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Glioma/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Animals , Casein Kinase II/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Glioma/pathology , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Phosphorylation/drug effects , Phosphoserine/metabolism , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/metabolism
9.
Cell Rep ; 17(1): 165-178, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27681429

ABSTRACT

In cold-blooded vertebrates such as zebrafish, Müller glial cells (MGs) readily proliferate to replenish lost retinal neurons. In mammals, however, MGs lack regenerative capability as they do not spontaneously re-enter the cell cycle unless the retina is injured. Here, we show that gene transfer of ß-catenin in adult mouse retinas activates Wnt signaling and MG proliferation without retinal injury. Upstream of Wnt, deletion of GSK3ß stabilizes ß-catenin and activates MG proliferation. Downstream of Wnt, ß-catenin binds to the Lin28 promoter and activates transcription. Deletion of Lin28 abolishes ß-catenin-mediated effects on MG proliferation, and Lin28 gene transfer stimulates MG proliferation. We further demonstrate that let-7 miRNAs are critically involved in Wnt/Lin28-regulated MG proliferation. Intriguingly, a subset of cell-cycle-reactivated MGs express markers for amacrine cells. Together, these results reveal a key role of Wnt-Lin28-let7 miRNA signaling in regulating proliferation and neurogenic potential of MGs in the adult mammalian retina.


Subject(s)
Ependymoglial Cells/metabolism , Gene Expression Regulation , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Wnt Proteins/genetics , Amacrine Cells/cytology , Amacrine Cells/metabolism , Animals , Cell Cycle/genetics , Cell Differentiation , Cell Proliferation , Ependymoglial Cells/cytology , Glycogen Synthase Kinase 3 beta/deficiency , Glycogen Synthase Kinase 3 beta/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , RNA-Binding Proteins/metabolism , Signal Transduction , Transcription, Genetic , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
10.
Neuron ; 90(1): 86-100, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27021171

ABSTRACT

Simultaneous co-activation of neocortical neurons is likely critical for brain computations ranging from perception and motor control to memory and cognition. While co-activation of excitatory principal cells (PCs) during ongoing activity has been extensively studied, that of inhibitory interneurons (INs) has received little attention. Here, we show in vivo and in vitro that members of two non-overlapping neocortical IN populations, expressing somatostatin (SOM) or vasoactive intestinal peptide (VIP), are active as populations rather than individually. We demonstrate a variety of synergistic mechanisms, involving population-specific local excitation, GABAergic disinhibition and excitation through electrical coupling, which likely underlie IN population co-activity. Firing of a few SOM or VIP INs recruits additional members within the cell type via GABAergic and cholinergic mechanisms, thereby amplifying the output of the population as a whole. Our data suggest that IN populations work as cooperative units, thus generating an amplifying nonlinearity in their circuit output.


Subject(s)
Acetylcholine/metabolism , Interneurons/metabolism , Neocortex/metabolism , Neural Inhibition/physiology , Pyramidal Cells/metabolism , Somatostatin/metabolism , Vasoactive Intestinal Peptide/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , In Vitro Techniques , Interneurons/physiology , Mice , Neurons/metabolism , Patch-Clamp Techniques
11.
Elife ; 5: e11903, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26974342

ABSTRACT

Axons fail to regenerate after central nervous system (CNS) injury. Modulation of the PTEN/mTORC1 pathway in retinal ganglion cells (RGCs) promotes axon regeneration after optic nerve injury. Here, we report that AKT activation, downstream of Pten deletion, promotes axon regeneration and RGC survival. We further demonstrate that GSK3ß plays an indispensable role in mediating AKT-induced axon regeneration. Deletion or inactivation of GSK3ß promotes axon regeneration independently of the mTORC1 pathway, whereas constitutive activation of GSK3ß reduces AKT-induced axon regeneration. Importantly, we have identified eIF2Bε as a novel downstream effector of GSK3ß in regulating axon regeneration. Inactivation of eIF2Bε reduces both GSK3ß and AKT-mediated effects on axon regeneration. Constitutive activation of eIF2Bε is sufficient to promote axon regeneration. Our results reveal a key role of the AKT-GSK3ß-eIF2Bε signaling module in regulating axon regeneration in the adult mammalian CNS.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Multiprotein Complexes/metabolism , Oncogene Protein v-akt/metabolism , Optic Nerve Injuries/pathology , Regeneration , Retinal Ganglion Cells/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Axons/physiology , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL
12.
Elife ; 52016 Feb 05.
Article in English | MEDLINE | ID: mdl-26848828

ABSTRACT

Aberrant signaling through the Raf/MEK/ERK (ERK/MAPK) pathway causes pathology in a family of neurodevelopmental disorders known as 'RASopathies' and is implicated in autism pathogenesis. Here, we have determined the functions of ERK/MAPK signaling in developing neocortical excitatory neurons. Our data reveal a critical requirement for ERK/MAPK signaling in the morphological development and survival of large Ctip2(+) neurons in layer 5. Loss of Map2k1/2 (Mek1/2) led to deficits in corticospinal tract formation and subsequent corticospinal neuron apoptosis. ERK/MAPK hyperactivation also led to reduced corticospinal axon elongation, but was associated with enhanced arborization. ERK/MAPK signaling was dispensable for axonal outgrowth of layer 2/3 callosal neurons. However, Map2k1/2 deletion led to reduced expression of Arc and enhanced intrinsic excitability in both layers 2/3 and 5, in addition to imbalanced synaptic excitation and inhibition. These data demonstrate selective requirements for ERK/MAPK signaling in layer 5 circuit development and general effects on cortical pyramidal neuron excitability.


Subject(s)
MAP Kinase Signaling System , Neocortex/embryology , Neurons/physiology , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Mice, Transgenic , Neurogenesis
13.
Cell ; 162(4): 795-807, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26255772

ABSTRACT

Deletion of UBE3A causes the neurodevelopmental disorder Angelman syndrome (AS), while duplication or triplication of UBE3A is linked to autism. These genetic findings suggest that the ubiquitin ligase activity of UBE3A must be tightly maintained to promote normal brain development. Here, we found that protein kinase A (PKA) phosphorylates UBE3A in a region outside of the catalytic domain at residue T485 and inhibits UBE3A activity toward itself and other substrates. A de novo autism-linked missense mutation disrupts this phosphorylation site, causing enhanced UBE3A activity in vitro, enhanced substrate turnover in patient-derived cells, and excessive dendritic spine development in the brain. Our study identifies PKA as an upstream regulator of UBE3A activity and shows that an autism-linked mutation disrupts this phosphorylation control. Moreover, our findings implicate excessive UBE3A activity and the resulting synaptic dysfunction to autism pathogenesis.


Subject(s)
Angelman Syndrome/genetics , Autistic Disorder/genetics , Mutation, Missense , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/metabolism , Animals , Autistic Disorder/metabolism , Brain/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dendritic Spines/pathology , Embryo, Mammalian/metabolism , Enzyme Stability , Female , Humans , Mice, Inbred C57BL , Mutagenesis , Phosphorylation , Ubiquitin-Protein Ligases/metabolism
15.
Dev Cell ; 31(6): 677-89, 2014 Dec 22.
Article in English | MEDLINE | ID: mdl-25535916

ABSTRACT

Coordinated migration of distinct classes of neurons to appropriate positions leads to the formation of functional neuronal circuitry in the cerebral cortex. The two major classes of cortical neurons, interneurons and projection neurons, utilize distinctly different modes (radial versus tangential) and routes of migration to arrive at their final positions in the cerebral cortex. Here, we show that adenomatous polyposis coli (APC) modulates microtubule (MT) severing in interneurons to facilitate tangential mode of interneuron migration, but not the glial-guided, radial migration of projection neurons. APC regulates the stability and activity of the MT-severing protein p60-katanin in interneurons to promote the rapid remodeling of neuronal processes necessary for interneuron migration. These findings reveal how severing and restructuring of MTs facilitate distinct modes of neuronal migration necessary for laminar organization of neurons in the developing cerebral cortex.


Subject(s)
Adenosine Triphosphatases/metabolism , Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Gene Expression Regulation, Developmental , Interneurons/metabolism , Microtubules/metabolism , Neurons/physiology , Alleles , Animals , Cell Differentiation , Cell Movement , Cerebral Cortex/metabolism , Cytoskeleton/metabolism , Gene Deletion , Green Fluorescent Proteins/metabolism , Katanin , Mice , Mice, Transgenic , Microscopy, Fluorescence , Neurons/metabolism , Time Factors
16.
Elife ; 3: e02663, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25073924

ABSTRACT

GSK-3 is an essential mediator of several signaling pathways that regulate cortical development. We therefore created conditional mouse mutants lacking both GSK-3α and GSK-3ß in newly born cortical excitatory neurons. Gsk3-deleted neurons expressing upper layer markers exhibited striking migration failure in all areas of the cortex. Radial migration in hippocampus was similarly affected. In contrast, tangential migration was not grossly impaired after Gsk3 deletion in interneuron precursors. Gsk3-deleted neurons extended axons and developed dendritic arbors. However, the apical dendrite was frequently branched while basal dendrites exhibited abnormal orientation. GSK-3 regulation of migration in neurons was independent of Wnt/ß-catenin signaling. Importantly, phosphorylation of the migration mediator, DCX, at ser327, and phosphorylation of the semaphorin signaling mediator, CRMP-2, at Thr514 were markedly decreased. Our data demonstrate that GSK-3 signaling is essential for radial migration and dendritic orientation and suggest that GSK-3 mediates these effects by phosphorylating key microtubule regulatory proteins.DOI: http://dx.doi.org/10.7554/eLife.02663.001.


Subject(s)
Cell Movement , Cerebral Cortex/cytology , Dendrites/metabolism , Glycogen Synthase Kinase 3/metabolism , Signal Transduction , Animals , Doublecortin Domain Proteins , Doublecortin Protein , Gene Deletion , Glycogen Synthase Kinase 3 beta , Hippocampus/cytology , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Neuropeptides/metabolism , PTEN Phosphohydrolase/metabolism , Phosphorylation , Substrate Specificity , Wnt Signaling Pathway , beta Catenin/metabolism , cdc42 GTP-Binding Protein/metabolism
17.
Neuron ; 83(3): 558-71, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25043421

ABSTRACT

Defects in ependymal (E) cells, which line the ventricle and generate cerebrospinal fluid flow through ciliary beating, can cause hydrocephalus. Dishevelled genes (Dvls) are essential for Wnt signaling, and Dvl2 has been shown to localize to the rootlet of motile cilia. Using the hGFAP-Cre;Dvl1(-/-);2(flox/flox);3(+/-) mouse, we show that compound genetic ablation of Dvls causes hydrocephalus. In hGFAP-Cre;Dvl1(-/-);2(flox/flox);3(+/-) mutants, E cells differentiated normally, but the intracellular and intercellular rotational alignments of ependymal motile cilia were disrupted. As a consequence, the fluid flow generated by the hGFAP-Cre;Dvl1(-/-);2(flox/flox);3(+/-) E cells was significantly slower than that observed in control mice. Dvls were also required for the proper positioning of motile cilia on the apical surface. Tamoxifen-induced conditional removal of Dvls in adult mice also resulted in defects in intracellular rotational alignment and positioning of ependymal motile cilia. These results suggest that Dvls are continuously required for E cell planar polarity and may prevent hydrocephalus.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Polarity/physiology , Cilia/pathology , Ependyma/pathology , Hydrocephalus/etiology , Phosphoproteins/genetics , Signal Transduction/physiology , Animals , Cell Polarity/genetics , Cilia/genetics , Dishevelled Proteins , Hydrocephalus/genetics , Hydrocephalus/pathology , Mice , Mice, Transgenic , Signal Transduction/genetics
18.
Curr Biol ; 23(17): R720-3, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-24028951

ABSTRACT

The polarity proteins LKB1 and SAD-A/B are key regulators of axon specification in the developing cerebral cortex. Recent studies now show that this mechanism cannot be generalized to other classes of neurons: instead, SAD-A/B functions downstream of neurotrophin signaling in sensory neurons to mediate a later stage of axon development - arborization in the target field.


Subject(s)
Axons/metabolism , Neurotrophin 3/metabolism , Protein Serine-Threonine Kinases/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction/physiology , Animals , Humans
19.
Neuron ; 75(6): 1035-50, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22998872

ABSTRACT

We have defined functions of MEK in regulating gliogenesis in developing cerebral cortex using loss- and gain-of-function mouse genetics. Radial progenitors deficient in both Mek1 and Mek2 fail to transition to the gliogenic mode in late embryogenesis, and astrocyte and oligodendroglial precursors fail to appear. In exploring mechanisms, we found that the key cytokine-regulated gliogenic pathway is attenuated. Further, the Ets transcription family member Etv5/Erm is strongly regulated by MEK and Erm overexpression can rescue the gliogenic potential of Mek-deleted progenitors. Remarkably, Mek1/2-deleted mice surviving postnatally exhibit cortices almost devoid of astrocytes and oligodendroglia and exhibit neurodegeneration. Conversely, expression of constitutively active MEK1 leads to a major increase in numbers of astrocytes in the adult brain. We conclude that MEK is essential for acquisition of gliogenic competence by radial progenitors and that levels of MEK activity regulate gliogenesis in the developing cortex.


Subject(s)
Brain , Gene Expression Regulation, Developmental/genetics , MAP Kinase Kinase 1/deficiency , MAP Kinase Kinase 2/deficiency , Neuroglia/physiology , Age Factors , Animals , Animals, Newborn , Brain/cytology , Brain/embryology , Brain/growth & development , Cell Count , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Ciliary Neurotrophic Factor/pharmacology , DNA-Binding Proteins/metabolism , Electroporation , Embryo, Mammalian , Excitatory Amino Acid Transporter 1/metabolism , Eye Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/metabolism , Ki-67 Antigen/metabolism , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , PAX6 Transcription Factor , Paired Box Transcription Factors/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics , Stem Cells/drug effects , Stem Cells/physiology , Transcription Factors/metabolism
20.
Neuron ; 73(4): 623-6, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22365537

ABSTRACT

In this issue of Neuron, Napoli et al. (2012) demonstrate that elevated ERK/MAPK signaling in Schwann cells is a crucial trigger for Schwann cell dedifferentiation in vivo. Moreover, the authors show that dedifferentiated Schwann cells have the potential to coordinate much of the peripheral nerve response to injury.

SELECTION OF CITATIONS
SEARCH DETAIL
...