Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Development ; 150(16)2023 08 15.
Article in English | MEDLINE | ID: mdl-37519286

ABSTRACT

Here, we show that, in the developing spinal cord, after the early Wnt-mediated Tcf transcription activation that confers dorsal identity to neural stem cells, neurogenesis redirects ß-catenin from the adherens junctions to the nucleus to stimulate Tcf-dependent transcription in a Wnt-independent manner. This new ß-catenin activity regulates genes implicated in several aspects of contralateral axon growth, including axon guidance and adhesion. Using live imaging of ex-vivo chick neural tube, we showed that the nuclear accumulation of ß-catenin and the rise in Tcf-dependent transcription both initiate before the dismantling of the adherens junctions and remain during the axon elongation process. Notably, we demonstrated that ß-catenin activity in post-mitotic cells depends on TCF7L2 and is central to spinal commissural axon growth. Together, our results reveal Wnt-independent Tcf/ß-catenin regulation of genes that control the growth and guidance of commissural axons in chick spinal cord.


Subject(s)
Neural Stem Cells , beta Catenin , beta Catenin/metabolism , Adherens Junctions/metabolism , Signal Transduction/physiology , Neurogenesis/genetics
2.
Elife ; 112022 02 08.
Article in English | MEDLINE | ID: mdl-35133277

ABSTRACT

Adenylosuccinate lyase (ADSL) functions in de novo purine synthesis (DNPS) and the purine nucleotide cycle. ADSL deficiency (ADSLD) causes numerous neurodevelopmental pathologies, including microcephaly and autism spectrum disorder. ADSLD patients have normal serum purine nucleotide levels but exhibit accumulation of dephosphorylated ADSL substrates, S-Ado, and SAICAr, the latter being implicated in neurotoxic effects through unknown mechanisms. We examined the phenotypic effects of ADSL depletion in human cells and their relation to phenotypic outcomes. Using specific interventions to compensate for reduced purine levels or modulate SAICAr accumulation, we found that diminished AMP levels resulted in increased DNA damage signaling and cell cycle delays, while primary ciliogenesis was impaired specifically by loss of ADSL or administration of SAICAr. ADSL-deficient chicken and zebrafish embryos displayed impaired neurogenesis and microcephaly. Neuroprogenitor attrition in zebrafish embryos was rescued by pharmacological inhibition of DNPS, but not increased nucleotide concentration. Zebrafish also displayed phenotypes commonly linked to ciliopathies. Our results suggest that both reduced purine levels and impaired DNPS contribute to neurodevelopmental pathology in ADSLD and that defective ciliogenesis may influence the ADSLD phenotypic spectrum.


Subject(s)
Adenylosuccinate Lyase/deficiency , Adenylosuccinate Lyase/metabolism , Autistic Disorder/metabolism , Neurogenesis , Purine-Pyrimidine Metabolism, Inborn Errors/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Autism Spectrum Disorder/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line , Chickens/metabolism , Ciliopathies/metabolism , DNA Damage , Humans , Microcephaly/metabolism , Microtubule-Associated Proteins/metabolism , Phenotype , Phosphoproteins/metabolism , Purines/metabolism , Ribonucleotides/metabolism , Zebrafish/metabolism
3.
J Neurosci ; 41(32): 6836-6849, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34210778

ABSTRACT

Adult neural plasticity is an important and intriguing phenomenon in the brain, and adult hippocampal neurogenesis is directly involved in modulating neural plasticity by mechanisms that are only partially understood. We have performed gain-of-function and loss-of-function experiments to study Smad2, a transcription factor selected from genes that are demethylated after exercise through the analysis of an array of physical activity-induced factors, and their corresponding gene expression, and an efficient inducer of plasticity. In these studies, changes in cell number and morphology were analyzed in the hippocampal dentate gyrus (cell proliferation and survival, including regional distribution, and structural maturation/differentiation, including arborization, dendritic spines, and neurotransmitter-specific vesicles) of sedentary male mice, after evaluation in a battery of behavioral tests. As a result, we reveal a role for Smad2 in the balance of proliferation versus maturation of differentiating immature cells (Smad2 silencing increases both the proliferation and survival of cycling cells in the dentate granule cell layer), and in the plasticity of both newborn and mature neurons in mice (by decreasing dendritic arborization and dendritic spine number). Moreover, Smad2 silencing specifically compromises spatial learning in mice (through impairments of spatial tasks acquisition both in long-term learning and working memory). These data suggest that Smad2 participates in adult neural plasticity by influencing the proliferation and maturation of dentate gyrus neurons.SIGNIFICANCE STATEMENT Smad2 is one of the main components of the transforming growth factor-ß (TGF-ß) pathway. The commitment of cell fate in the nervous system is tightly coordinated by SMAD2 signaling, as are further differentiation steps (e.g., dendrite and axon growth, myelination, and synapse formation). However, there are no studies that have directly evaluated the role of Smad2 gene in hippocampus of adult animals. Modulation of these parameters in the adult hippocampus can affect hippocampal-dependent behaviors, which may shed light on the mechanisms that regulate adult neurogenesis and behavior. We demonstrate here a role for Smad2 in the maturation of differentiating immature cells and in the plasticity of mature neurons. Moreover, Smad2 silencing specifically compromises the spatial learning abilities of adult male mice.


Subject(s)
Dentate Gyrus/physiology , Neuronal Plasticity/physiology , Smad2 Protein/metabolism , Spatial Learning/physiology , Spatial Memory/physiology , Animals , Male , Mice , Mice, Inbred C57BL , Neurogenesis/physiology
4.
J Cell Biol ; 220(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-33939796

ABSTRACT

The neural tube forms when neural stem cells arrange into a pseudostratified, single-cell-layered epithelium, with a marked apico-basal polarity, and in which adherens junctions (AJs) concentrate in the subapical domain. We previously reported that sustained ß-catenin expression promotes the formation of enlarged apical complexes (ACs), enhancing apico-basal polarity, although the mechanism through which this occurs remained unclear. Here, we show that ß-catenin interacts with phosphorylated pro-N-cadherin early in its transit through the Golgi apparatus, promoting propeptide excision and the final maturation of N-cadherin. We describe a new ß-catenin-dependent interaction of N-cadherin with Drebrin-like (Dbnl), an actin-binding protein that is involved in anterograde Golgi trafficking of proteins. Notably, Dbnl knockdown led to pro-N-cadherin accumulation and limited AJ formation. In brief, we demonstrate that Dbnl and Drebrin-like ß-catenin assist in the maturation of pro-N-cadherin, which is critical for AJ formation and for the recruitment AC components like aPKC and, consequently, for the maintenance of apico-basal polarity.


Subject(s)
Adherens Junctions/physiology , Antigens, CD/metabolism , Cadherins/metabolism , Cell Polarity , Microfilament Proteins/metabolism , Neural Stem Cells/metabolism , beta Catenin/metabolism , Antigens, CD/genetics , Cadherins/genetics , Cells, Cultured , HEK293 Cells , Humans , Microfilament Proteins/genetics , Neural Stem Cells/cytology , beta Catenin/genetics , src Homology Domains/genetics
5.
Bioorg Chem ; 108: 104660, 2021 03.
Article in English | MEDLINE | ID: mdl-33550073

ABSTRACT

A structure-activity relationship (SAR) study in terms of G-quadruplex binding ability and antiproliferative activity of six fluorescent perylenemonoimide (PMIs) derivatives is reported. A positive charge seems to be the key to target G4. This study also reveals the importance of the element substitution in the potential biological activity of PMIs, being the polyethylene glycol (PEG) chains in the peri position responsible for their antiproliferative activity. Among them, the cationic PMI6 with two PEG chains is the most promising compound since its fluorescence is enhanced in the presence of G-quadruplex structures. Moreover, PMI6 binds to the human telomeric G-quadruplex hTelo with high affinity and displays a high antiproliferative potential towards HeLa (cervical adenocarcinoma), A549 (lung adenocarcinoma) and A2780 (ovarian adenocarcinoma) cells. Its fate can be followed inside cells thanks to its fluorescent properties: the compound is found to accumulate in the mitochondria.


Subject(s)
G-Quadruplexes/drug effects , Imides/pharmacology , Perylene/analogs & derivatives , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Imides/chemical synthesis , Imides/chemistry , Mitochondria/drug effects , Molecular Structure , Perylene/chemical synthesis , Perylene/chemistry , Perylene/pharmacology , Structure-Activity Relationship
6.
Elife ; 72018 08 10.
Article in English | MEDLINE | ID: mdl-30095408

ABSTRACT

Class II HLH proteins heterodimerize with class I HLH/E proteins to regulate transcription. Here, we show that E proteins sharpen neurogenesis by adjusting the neurogenic strength of the distinct proneural proteins. We find that inhibiting BMP signaling or its target ID2 in the chick embryo spinal cord, impairs the neuronal production from progenitors expressing ATOH1/ASCL1, but less severely that from progenitors expressing NEUROG1/2/PTF1a. We show this context-dependent response to result from the differential modulation of proneural proteins' activity by E proteins. E proteins synergize with proneural proteins when acting on CAGSTG motifs, thereby facilitating the activity of ASCL1/ATOH1 which preferentially bind to such motifs. Conversely, E proteins restrict the neurogenic strength of NEUROG1/2 by directly inhibiting their preferential binding to CADATG motifs. Since we find this mechanism to be conserved in corticogenesis, we propose this differential co-operation of E proteins with proneural proteins as a novel though general feature of their mechanism of action.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA/metabolism , Gene Expression Regulation, Developmental , Neurogenesis , Animals , Binding Sites , Chick Embryo , Protein Binding
7.
Dev Biol ; 436(1): 42-54, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29470955

ABSTRACT

Phosphatidylinositol 3-kinases (PI3Ks) are signal transducers of many biological processes. Class 1 A PI3Ks are hetero dimers formed by a regulatory and a catalytic subunit. We have used the developing chicken neural tube (NT) to study the roles played by PI3K during the process of cell proliferation and differentiation. Notably, we have observed that in addition to its well characterized anti apoptotic activity, PI3K also plays a crucial role in intra epithelial cell positioning, and unlike its role in survival that mainly depends on AKT, the activity in cell positioning is mediated by Rho GTPase family members. Additionally, we have observed that activating mutations of PI3K that are remarkably frequent in many human cancers, cause an unrestrained basal migration of the neuroepithelial cells that end up breaking through the basal membrane invading the surrounding mesenchymal tissue. The mechanism described in this work contribute not only to acquire a greater knowledge of the intraepithelial cell positioning process, but also give new clues on how activating mutations of PI3K contribute to cell invasion during the first stages of tumour dissemination.


Subject(s)
Cell Polarity/genetics , Neural Tube/metabolism , Neurogenesis/genetics , Phosphatidylinositol 3-Kinases/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , Chick Embryo , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Neural Tube/embryology , Neuroepithelial Cells/metabolism , Neurogenesis/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
8.
Development ; 143(12): 2194-205, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27122165

ABSTRACT

Delamination of neural crest (NC) cells is a bona fide physiological model of epithelial-to-mesenchymal transition (EMT), a process that is influenced by Wnt/ß-catenin signalling. Using two in vivo models, we show that Wnt/ß-catenin signalling is transiently inhibited at the time of NC delamination. In attempting to define the mechanism underlying this inhibition, we found that the scaffold proteins Dact1 and Dact2, which are expressed in pre-migratory NC cells, are required for NC delamination in Xenopus and chick embryos, whereas they do not affect the motile properties of migratory NC cells. Dact1/2 inhibit Wnt/ß-catenin signalling upstream of the transcriptional activity of T cell factor (TCF), which is required for EMT to proceed. Dact1/2 regulate the subcellular distribution of ß-catenin, preventing ß-catenin from acting as a transcriptional co-activator to TCF, yet without affecting its stability. Together, these data identify a novel yet important regulatory element that inhibits ß-catenin signalling, which then affects NC delamination.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Wnt Proteins/metabolism , Animals , Cell Movement , Cell Nucleus/metabolism , Chick Embryo , Female , HEK293 Cells , Humans , Subcellular Fractions/metabolism , Wnt Signaling Pathway , Xenopus laevis/embryology , Xenopus laevis/metabolism , beta Catenin/metabolism
9.
J Cell Sci ; 128(15): 2928-37, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26092933

ABSTRACT

Protein kinase A (PKA) accumulates at the base of the cilium where it negatively regulates the Hedgehog (Hh) pathway. Although PKA activity is essentially controlled by the cAMP produced by adenylyl cyclases, the influence of these enzymes on the Hh pathway remains unclear. Here, we show that adenylyl cyclase 5 and adenylyl cyclase 6 (AC5 and AC6, also known as ADCY5 and ADCY6, respectively) are the two isoforms most strongly expressed in cerebellar granular neuron precursors (CGNPs). We found that overexpression of AC5 and AC6 represses, whereas their knockdown activates, the Hh pathway in CGNPs and in the embryonic neural tube. Indeed, AC5 and AC6 concentrate in the primary cilium, and mutation of a previously undescribed cilium-targeting motif in AC5 suppresses its ciliary location, as well as its capacity to inhibit Hh signalling. Stimulatory and inhibitory Gα proteins, which are engaged by the G-protein-coupled receptors (GPCRs), control AC5 and AC6 activity and regulate the Hh pathway in CGNPs and in the neural tube. Therefore, we propose that the activity of different ciliary GPCRs converges on AC5 and AC6 to control PKA activity and, hence, the Hh pathway.


Subject(s)
Adenylyl Cyclases/genetics , Cilia/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Hedgehog Proteins/metabolism , 3T3 Cells , Adenylyl Cyclases/metabolism , Animals , Cell Line , Cell Proliferation/genetics , Chick Embryo , GTP-Binding Protein alpha Subunits/genetics , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Tube/cytology , Neural Tube/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction/physiology , Zinc Finger Protein Gli2 , Zinc Finger Protein Gli3
10.
Chem Sci ; 6(7): 3757-3764, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-29218145

ABSTRACT

Selected new fluorogenic probes that interact in different ways with Hg2+ and MeHg+ have been prepared and used for the chemical speciation of both cations in aqueous solution as well as in HEK293 cells. The best selective speciation of Hg2+ and MeHg+ has been achieved by in vitro approaches based on fluorogenic probes supported in cultured cells, due to the particular sensitivity of the HEK293 cells to permeation by Hg2+, MeHg+ and the fluorogenic probes. In particular, MeHg+ was selectively detected in cell nuclei by probe JG45.

11.
Nat Commun ; 5: 4168, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24942669

ABSTRACT

ß-Catenin mediates the canonical Wnt pathway by stimulating Tcf-dependent transcription and also associates to N-cadherin at the apical complex (AC) of neuroblasts. Here, we show that while ß-catenin activity is required to form the AC and to maintain the cell polarity, oncogenic mutations that render stable forms of ß-catenin (sß-catenin) maintain the stemness of neuroblasts, inhibiting their differentiation and provoking aberrant growth. In examining the transcriptional and structural roles of ß-catenin, we find that while ß-catenin/Tcf transcriptional activity induces atypical protein kinase C (aPKC) expression, an alternative effect of ß-catenin restricts aPKC to the apical pole of neuroepithelial cells. In agreement, we show that a constitutively active form of aPKC reproduces the neuroepithelial aberrations induced by ß-catenin. Therefore, we conclude that ß-catenin controls the cell fate and polarity of the neuroblasts through the expression and localization of aPKC.


Subject(s)
Cell Polarity , Chickens/metabolism , Epithelial Cells/cytology , Neurons/cytology , Protein Kinase C/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Chick Embryo , Chickens/genetics , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Neurons/enzymology , Neurons/metabolism , Protein Kinase C/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway , beta Catenin/genetics
12.
J Cell Sci ; 126(Pt 23): 5335-43, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24105267

ABSTRACT

The transforming growth factor beta (TGF-ß) pathway plays key roles in development and cancer. TGF-ß signaling converges on the Smad2 and Smad3 effectors, which can either cooperate or antagonize to regulate their transcriptional targets. Here we performed in vivo and in silico experiments to study how such cooperativity and antagonism might function during neurogenesis. In vivo electroporation experiments in the chick embryo neural tube show that Smad2 and Smad3 cooperate to promote neurogenesis, as well as the transcription of Smad3-specific targets. Knockdown of Smad2 enhances neurogenesis and the transcription of Smad3-specific targets. A mathematical model of the TGF-ß pathway fits the experimental results and predicts that the proportions of the three different trimeric complexes formed dictates the transcriptional responses of the R-Smad proteins. As such, Smad2 targets are activated solely by the Smad2-Smad2-Smad4 complex, whereas Smad3 targets are activated both by Smad2-Smad3-Smad4 and Smad3-Smad3-Smad4 trimers. We have modeled the Smad responses onto arbitrary genes and propose that this mechanism might be extended to additional activities of TGF-ß in development and disease.


Subject(s)
Gene Expression Regulation, Developmental , Neurogenesis/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Smad4 Protein/genetics , Animals , Chick Embryo , Computer Simulation , Electroporation , Models, Genetic , Protein Multimerization , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Smad2 Protein/antagonists & inhibitors , Smad2 Protein/metabolism , Smad3 Protein/antagonists & inhibitors , Smad3 Protein/metabolism , Smad4 Protein/antagonists & inhibitors , Smad4 Protein/metabolism , Transcription, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
13.
Mol Cell Biol ; 33(14): 2706-17, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23671190

ABSTRACT

During cerebellum development, Sonic hedgehog (Shh)-induced proliferation of cerebellar granular neuronal precursors (CGNPs) is potently inhibited by bone morphogenetic proteins (BMPs). We have previously reported the upregulation of TIEG-1 and Mash1, two antimitotic factors that modulate MYCN transcription and N-Myc activity, in response to BMP2. To gain further insight into the BMP antimitotic mechanism, we used microRNA (miRNA) arrays to compare the miRNAs of CGNPs proliferating in response to Shh with those of CGNPs treated with Shh plus BMP2. The array analysis revealed that miRNA 11 (miR-22) levels significantly increased in cells treated with BMP2. Additionally, in P7 mouse cerebellum, miR-22 distribution mostly recapitulated the combination of BMP2 and BMP4 expression patterns. Accordingly, in CGNP cultures, miR-22 overexpression significantly reduced cell proliferation, whereas miR-22 suppression diminished BMP2 antiproliferative activity. In contrast to BMP2, miR-22 did not induce neural differentiation but instead significantly increased cell cycle length. Consistent with the central role played by N-myc on CGNP proliferation, Max was revealed as a direct target of miR-22, and miR-22 expression caused a significant reduction of Max protein levels and N-myc/Max-dependent promoter activity. Therefore, we conclude that, in addition to the previously described mechanisms, miR-22 plays a specific role on downstream BMPs through cerebellum growth.


Subject(s)
Cell Cycle Checkpoints/genetics , Cerebellum/cytology , MicroRNAs/physiology , Neural Stem Cells/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bone Morphogenetic Proteins/physiology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Mice , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Signal Transduction , Transcription, Genetic , Transcriptome
14.
Cereb Cortex ; 22(8): 1717-27, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21955917

ABSTRACT

The beneficial effects of insulin and insulin-like growth factor I on cognition have been documented in humans and animal models. Conversely, obesity, hyperinsulinemia, and diabetes increase the risk for neurodegenerative disorders including Alzheimer's disease (AD). However, the mechanisms by which insulin regulates synaptic plasticity are not well understood. Here, we report that complete disruption of insulin receptor substrate 2 (Irs2) in mice impairs long-term potentiation (LTP) of synaptic transmission in the hippocampus. Basal synaptic transmission and paired-pulse facilitation were similar between the 2 groups of mice. Induction of LTP by high-frequency conditioning tetanus did not activate postsynaptic N-methyl-D-aspartate (NMDA) receptors in hippocampus slices from Irs2(-/-) mice, although the expression of NR2A, NR2B, and PSD95 was equivalent to wild-type controls. Activation of Fyn, AKT, and MAPK in response to tetanus stimulation was defective in Irs2(-/-) mice. Interestingly, IRS2 was phosphorylated during induction of LTP in control mice, revealing a potential new component of the signaling machinery which modulates synaptic plasticity. Given that IRS2 expression is diminished in Type 2 diabetics as well as in AD patients, these data may reveal an explanation for the prevalence of cognitive decline in humans with metabolic disorders by providing a mechanistic link between insulin resistance and impaired synaptic transmission.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , Long-Term Potentiation/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , Animals , Blotting, Western , Female , Hippocampus/metabolism , Immunoprecipitation , Insulin Receptor Substrate Proteins/deficiency , Mice , Mice, Knockout , Patch-Clamp Techniques
15.
J Biol Chem ; 286(10): 8067-8074, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21209076

ABSTRACT

Proliferation of cerebellar granular neuronal precursors (CGNPs) is mediated by Sonic Hedgehog (Shh), which activates the Patched and Smoothened (Smo) receptor complex. Although its protein sequence suggests that Smo is a G protein coupled receptor (GPCR), the evidence that this receptor utilizes heterotrimeric G proteins as downstream effectors is controversial. In Drosophila, Gα(i) is required for Hedgehog (Hh) activity, but the involvement of heterotrimeric G proteins in vertebrate Shh signaling has not yet been established. Here, we show that Shh-induced proliferation of rat CGNPs is enhanced strongly by the expression of the active forms of Gα(i/o) proteins (Gα(i1), Gα(i2), Gα(i3), and Gα(o)) but not by members of another class (Gα(12)) of heterotrimeric G proteins. Additionally, the mRNAs of these different Gα(i) members display specific expression patterns in the developing cerebellum; only Gα(i2) and Gα(i3) are substantially expressed in the outer external granular layer, where CGNPs proliferate. Consistent with this, Shh-induced proliferation of CGNPs is reduced significantly by knockdowns of Gα(i2) and Gα(i3) but not by silencing of other members of the Gα(i/o) class. Finally, our results demonstrate that Gα(i2) and Gα(i3) locate to the primary cilium when expressed in CGNP cultures. In summary, we conclude that the proliferative effects of Shh on CGNPs are mediated by the combined activity of Gα(i2) and Gα(i3) proteins.


Subject(s)
Cell Proliferation , Cerebellum/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Hedgehog Proteins/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Cells, Cultured , Cerebellum/cytology , Drosophila melanogaster , GTP-Binding Protein alpha Subunits/genetics , Neurons/cytology , Rats , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Smoothened Receptor , Stem Cells/cytology
17.
J Cell Sci ; 123(Pt 1): 62-9, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20016067

ABSTRACT

Cerebellar granular neuronal precursors (CGNPs) proliferate in response to the mitogenic activity of Sonic hedgehog (Shh), and this proliferation is negatively regulated by activation of cAMP-dependent protein kinase (PKA). In the basal state, the PKA catalytic subunits (C-PKA) are inactive because of their association with the regulatory subunits (R-PKA). As the level of cAMP increases, it binds to R-PKA, displacing and thereby activating the C-PKA. Here we report that, in the presence of Shh, inactive C-PKA accumulates at the cilium base of proliferative CGNPs whereas removal of Shh triggers the activation of PKA at this particular location. Furthermore, we demonstrate that the anchoring of the PKA holoenzyme to the cilium base is mediated by the specific binding of the type II PKA regulatory subunit (RII-PKA) to the A-kinase anchoring proteins (AKAPs). Disruption of the interaction between RII-PKA and AKAPs inhibits Shh activity and, therefore, blocks proliferation of CGNP cultures. Collectively, these results demonstrate that the pool of PKA localized to the cilium base of CGNP plays an essential role in the integration of Shh signal transduction.


Subject(s)
Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Hedgehog Proteins/metabolism , Protein Subunits/metabolism , Stem Cells/metabolism , A Kinase Anchor Proteins/metabolism , Animals , Cell Proliferation , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cilia , Enzyme Activation , Mice , Protein Binding , Protein Transport , Signal Transduction , Stem Cells/cytology
18.
Toxicol Sci ; 112(2): 416-26, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19770487

ABSTRACT

Methylmercury (MeHg) is an environmental neurotoxicant whose molecular mechanisms underlying toxicity remain elusive. Here, we investigated molecular events involved in MeHg-induced neurotoxicity in cultured cerebellar granule cells (CGCs) as well as potential protective strategies for such toxicity. Glutathione peroxidase, isozyme 1 (GPx-1) activity was significantly (p = 0.0017) decreased at 24 h before MeHg-induced neuronal death (day in vitro 4). This event was related to enhanced susceptibilities to hydrogen peroxide or tert-butyl peroxide and increased lipid peroxidation. However, intracellular calcium levels, glutamate uptake, and glutathione levels, as well as glutathione reductase and catalase activities, were not changed by MeHg exposure at this time point. Probucol (PB), a lipid-lowering drug, displayed a long-lasting protective effect against MeHg-induced neurotoxicity. The beneficial effects of PB were correlated with increased GPx-1 activity and decreased lipid peroxidation. The protection afforded by PB was significantly higher when compared to the antioxidants, ascorbic acid and trolox. In vitro studies with the purified GPx-1 proved that MeHg inhibits and PB activates the enzyme activity. Overexpression of GPx-1 prevented MeHg-induced neuronal death. These data indicate that (1) GPx-1 is an important molecular target involved in MeHg-induced neurotoxicity and (2) PB, which increases GPx-1 activity in CGCs, induces enduring protection against such toxicity. The results bring out new insights on the potential therapeutic strategies for poisonings to MeHg and other pathological conditions related to increased production and/or decreased detoxification of peroxides.


Subject(s)
Anticholesteremic Agents/pharmacology , Cerebellum/drug effects , Cytoplasmic Granules/drug effects , Glutathione Peroxidase/metabolism , Methylmercury Compounds/toxicity , Probucol/pharmacology , Animals , Antioxidants/pharmacology , Catalase/metabolism , Cerebellum/metabolism , Cytoplasmic Granules/metabolism , Enzyme Activation , Glutathione/metabolism , Glutathione Reductase/metabolism , Lipid Peroxidation , Mice , Glutathione Peroxidase GPX1
19.
J Cell Sci ; 122(Pt 5): 595-9, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19208763

ABSTRACT

Murine Mash1 (Ascl1) is a member of the basic helix-loop-helix family of transcription factors and has been described to promote differentiation in some neural precursors. The process of differentiation is coordinated with a concomitant cell-cycle arrest, but the molecular mechanism of this process is unclear. Here, we describe for the very first time a direct regulation of an oncogene by a proneural gene. When expressed in proliferating cerebellar granular precursors, expression of the proneural gene encoding Mash1 promotes cell-cycle exit and differentiation, whereas expression of the oncogene MYCN has the opposite effect, promoting the proliferation of these cells in the absence of sonic hedgehog. Moreover, Mash1 overexpression neutralizes MYCN-induced proliferation. We now propose that the mechanism of antagonism between both molecules is based on opposite functions in the transcriptional regulation of the E-box motif, particularly in the E-boxes within the cyclin-D2 promoter, with MYCN acting as a transcriptional activator and Mash1 as a repressor. In agreement with this result, overexpression of cyclin D2 suppressed the anti-proliferative activity of Mash1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation , Mitosis/physiology , Neurons/physiology , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebellum/cytology , Cyclin D2 , Cyclins/genetics , Cyclins/metabolism , Humans , Mice , N-Myc Proto-Oncogene Protein , Neurons/cytology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Promoter Regions, Genetic , Stem Cells/cytology , Stem Cells/physiology
20.
J Biol Chem ; 282(51): 37170-80, 2007 Dec 21.
Article in English | MEDLINE | ID: mdl-17951258

ABSTRACT

Nmyc is a potent regulator of cell cycle in cerebellar granular neuron precursors (CGNPs) and has been proposed to be the main effector of Shh (Sonic hedgehog) proliferative activity. Nmyc ectopic expression is sufficient to promote cell autonomous proliferation and can lead to tumorigenesis. Bone morphogenetic protein 2 (BMP2) antagonizes Shh proliferative effect by promoting cell cycle exit and differentiation in CGNPs. Here we report that BMP2 opposes Shh mitogenic activity by blocking Nmyc expression. We have identified TIEG-1 (KLF10) as the intermediary factor that blocks Nmyc expression through the occupancy of the Sp1 sites present in its promoter. We also demonstrate that TIEG-1 ectopic expression in CGNPs induces cell cycle arrest that can lead to apoptosis but fails to promote differentiation. Moreover, TIEG-1 synergizes with BMP2 activity to terminally differentiate CGNPs and independent differentiator signals such as dibutyryl cAMP and prevents apoptosis in TIEG-1 arrested cells. All together, these data strongly suggest that the BMP2 pathway triggers cell cycle exit and differentiation as two separated but coordinated processes, where TIEG-1 acts as a mediator of the cell cycle arrest.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Cycle/physiology , Cerebellum/metabolism , DNA-Binding Proteins/metabolism , Hedgehog Proteins/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/genetics , Bucladesine/pharmacology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Cerebellum/cytology , DNA-Binding Proteins/genetics , Gene Expression , Hedgehog Proteins/genetics , Mice , Neurons/cytology , Proto-Oncogene Proteins c-myc/genetics , Response Elements/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/cytology , Transcription Factors/genetics , Transforming Growth Factor beta/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...