Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 328
1.
Toxicology ; 465: 153052, 2022 01 15.
Article En | MEDLINE | ID: mdl-34838597

Bisphenol-A (BPA), an environmental endocrine disruptor, is toxic to the central nervous system. Although recent studies have shown BPA-induced neurotoxicity, it is far from clear what precisely epigenetic mechanisms are involved in BPA-induced cognitive deficits. In this study, pheochromocytoma (PC12) cells were treated with BPA at 1 µM for 36 h in vitro. In vivo, C57BL/6 mice were administered to BPA at a dose of 1 mg/kg/day for 10 weeks. The results showed that 1 µM BPA exposure for 36 h impaired neurite outgrowth of PC12 cells through decreasing the primary and secondary branches. Besides, BPA exposure decreased the level of Ac-H3K9 (histone H3 Lys9 acetylation) by upregulating the expression of HDAC2 (histone deacetylases 2) in PC12 cells. Furthermore, treatment of both TSA (Trichostatin A, inhibitor of the histone deacetylase) and shHDAC2 plasmid (HDAC2 knockdown construct) resulted in amelioration neurite outgrowth deficits induced by BPA. In addition, it was shown that repression of HDAC2 could markedly rescue the spine density impairment in the hippocampus and prevent the cognitive impairment caused by BPA exposure in mice. Collectively, HDAC2 plays an essential role in BPA-induced neurotoxicity, which provides a potential molecular target for medical intervention.


Benzhydryl Compounds/toxicity , Dendritic Spines/drug effects , Environmental Pollutants/toxicity , Hippocampus/drug effects , Histone Deacetylase 2/metabolism , Neurites/drug effects , Neurotoxicity Syndromes/etiology , Phenols/toxicity , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Dendritic Spines/enzymology , Dendritic Spines/pathology , Female , Hippocampus/enzymology , Hippocampus/pathology , Hippocampus/physiopathology , Histone Deacetylase 2/genetics , Male , Maze Learning/drug effects , Mice, Inbred C57BL , Neurites/enzymology , Neurites/pathology , Neuronal Outgrowth/drug effects , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/physiopathology , PC12 Cells , Rats , Up-Regulation
2.
Neurochem Int ; 139: 104795, 2020 10.
Article En | MEDLINE | ID: mdl-32652266

Collapsin response mediator proteins (CRMPs) are ubiquitously expressed in neurons from worms to humans. A cardinal feature of CRMPs is to mediate growth cone collapse in response to Semaphorin-3A signaling through interactions with cytoskeletal proteins. These are critical regulatory roles that CRMPs play during neuritogenesis and neural network formation. Through post-translational modifications, such as phosphorylation, O-GlcNAcylation, SUMOylation, and proteolytic cleavage, CRMPs participate in synaptic plasticity by modulating NMDA receptors, L- and N-type voltage-gated calcium channels (VGCCs), thus affecting neurotransmitter release. CRMPs also possess histone deacetylase (HDAC) activity, which deacetylates histone H4 during neuronal death. Calcium-dependent proteolytic cleavage of CRMPs results in the truncation of CRMPs, producing a large 54 kD fragment (p54). Translocation of the p54 fragment into the nucleus leads to deacetylation of nuclear histone H4 and de-repression of transcription factor E2F1 expression. Increased expression of E2F1 elevates the expression of genes in cell cycle and death. These new and exciting studies lead to the realization that CRMPs are multifunctional proteins with both regulatory and enzymatic functions. Increasing numbers of studies associate these functions of CRMPs with the development of mental and neurological disorders, such as schizophrenia, Alzheimer's diseases, brain trauma, and stroke. This review focuses on new evidence showing the regulatory and enzymatic functions of CRMPs and highlights recent understandings of CRMPs' roles in neurological diseases.


Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurites/enzymology , Neuronal Plasticity/physiology , Transcription, Genetic/physiology , Animals , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Neurites/pathology
3.
Elife ; 92020 05 07.
Article En | MEDLINE | ID: mdl-32378514

Structural changes in pre and postsynaptic neurons that accompany synapse formation often temporally and spatially overlap. Thus, it has been difficult to resolve which processes drive patterned connectivity. To overcome this, we use the laminated outer murine retina. We identify the serine/threonine kinase LKB1 as a key driver of synapse layer emergence. The absence of LKB1 in the retina caused a marked mislocalization and delay in synapse layer formation. In parallel, LKB1 modulated postsynaptic horizontal cell refinement and presynaptic photoreceptor axon growth. Mislocalized horizontal cell processes contacted aberrant cone axons in LKB1 mutants. These defects coincided with altered synapse protein organization, and horizontal cell neurites were misdirected to ectopic synapse protein regions. Together, these data suggest that LKB1 instructs the timing and location of connectivity in the outer retina via coordinate regulation of pre and postsynaptic neuron structure and the localization of synapse-associated proteins.


Neurites/enzymology , Neurogenesis , Photoreceptor Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Synapses/enzymology , AMP-Activated Protein Kinases , Animals , Female , Male , Mice, Knockout , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Transport , Vesicular Glutamate Transport Protein 1/metabolism
4.
Brain Res Bull ; 142: 156-162, 2018 09.
Article En | MEDLINE | ID: mdl-30016727

Conventional protein kinase C (cPKC)γ and synapsin Ia/b have been implicated in the development of ischemic stroke, but their relationships and functions are unclear. In the present study, the oxygen-glucose deprivation (OGD)-induced ischemic insult in primary cultured cortical neurons in vitro and middle cerebral artery occlusion (MCAO)-induced ischemic stroke model in vivo were used to elucidate the function of cPKCγ and its modulation on synapsin Ia/b phosphorylation in ischemic stroke. We found that cPKCγ knockout significantly increased the infarct volume of mice after 1 h MCAO/72 h reperfusion by using triphenyltetrazolium chloride (TTC) staining. In the primarily cultured cortical neurons, cPKCγ knockout also aggravated the OGD-induced cell death and morphological damage of neurites, while cPKCγ restoration could alleviate the ischemic injury. Among the five phosphorylation sites of synapsin Ia/b, only the phosphorylation levels of Ser549 and 553 could be modulated by cPKCγ in neurons following 0.5 h OGD/24 h reoxygenation. In addition, we found that cPKCγ and synapsin Ia/b could be reciprocally co-immunoprecipitated in the cerebral cortex of MCAO mice. Taken together, we proposed that cPKCγ alleviates ischemic injury through modulating Ser549/553- synapsin Ia/b phosphorylation in neurons of mice.


Brain Ischemia/enzymology , Cell Hypoxia/physiology , Neurons/enzymology , Protein Kinase C/deficiency , Reperfusion Injury/enzymology , Synapsins/metabolism , Acyltransferases/physiology , Animals , Brain Ischemia/pathology , Cells, Cultured , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Models, Animal , Escherichia coli Proteins/physiology , Glucose/deficiency , Male , Mice, Inbred C57BL , Mice, Knockout , Neurites/enzymology , Neurites/pathology , Neurons/pathology , Neuroprotection/physiology , Primary Cell Culture , Protein Kinase C/genetics , Reperfusion Injury/pathology , Stroke/enzymology , Stroke/pathology
5.
eNeuro ; 5(1)2018.
Article En | MEDLINE | ID: mdl-29497702

Damage to the CNS results in neuronal and axonal degeneration, and subsequent neurological dysfunction. Endogenous repair in the CNS is impeded by inhibitory chemical and physical barriers, such as chondroitin sulfate proteoglycans (CSPGs) and myelin-associated glycoprotein (MAG), which prevent axon regeneration. Previously, it has been demonstrated that the inhibition of axonal histone deacetylase-6 (HDAC6) can promote microtubule α-tubulin acetylation and restore the growth of CSPGs- and MAG-inhibited axons. Since the acetylation of α-tubulin is regulated by two opposing enzymes, HDAC6 (deacetylation) and α-tubulin acetyltransferase-1 (αTAT1; acetylation), we have investigated the regulation of these enzymes downstream of a growth inhibitory signal. Our findings show that exposure of primary mouse cortical neurons to soluble CSPGs and MAG substrates cause an acute and RhoA-kinase-dependent reduction in α-tubulin acetylation and αTAT1 protein levels, without changes to either HDAC6 levels or HDAC6 activity. The CSPGs- and MAG-induced reduction in αTAT1 occurs primarily in the distal and middle regions of neurites and reconstitution of αTAT1, either by Rho-associated kinase (ROCK) inhibition or lentiviral-mediated αTAT1 overexpression, can restore neurite growth. Lastly, we demonstrate that CSPGs and MAG signaling decreases αTAT1 levels posttranscriptionally via a ROCK-dependent increase in αTAT1 protein turnover. Together, these findings define αTAT1 as a novel potential therapeutic target for ameliorating CNS injury characterized by growth inhibitory substrates that are prohibitive to axonal regeneration.


Acetyltransferases/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Myelin-Associated Glycoprotein/metabolism , Nerve Regeneration , Neurites/enzymology , Neuronal Outgrowth , Tubulin/metabolism , Animals , Down-Regulation , Female , Histone Deacetylase 6/metabolism , Mice , Microtubule Proteins/metabolism , Signal Transduction , rho-Associated Kinases/metabolism
6.
FASEB J ; 32(5): 2891-2898, 2018 05.
Article En | MEDLINE | ID: mdl-29401590

Cells have developed lineage-specific mechanisms to control proliferation and drive morphologic changes upon differentiation. A hallmark of differentiation is the assembly of signaling molecules that transduce extracellular signals, such as the production of the G protein-regulated enzyme phospholipase Cß (PLCß), which generates calcium signals from sensory stimuli. We found that in most cancerous cell lines there is positive correlation between PLCß1 levels and cell proliferation. In cells of neuronal lineage, however, reducing PLCß1 levels increases the rate of proliferation. Using a combination of biochemical and biophysical methods, we find that, in the G1 phase, a cytosolic population of PLCß1 associates with cyclin-dependent kinase 16 (CDK16), a neuron-specific enzyme that is activated by cyclin Y to inactivate the antioncogenic protein p27Kip1. Binding of PLCß1 directly inhibits CDK16 activity and in turn reduces the ability of cells to enter the S phase. Activation of Gαq by carbachol causes movement of PLCß from the cytosol to the plasma membrane, reducing its association with CDK16. Similarly, the overexpression of activated Gαq moves PLCß1 to the membrane, reverses G1 arrest, and promotes proliferation, thereby connecting external stimuli with cell proliferation. Our results present a model in which the transient high expression of PLCß1 that occurs at the onset of differentiation arrests cells in the G1 phase through its association with CDK16 and allows CDK16 to transition to its postmitotic function of neurite outgrowth and trafficking of synaptic vesicles. The novel role of PLCß1 in neuronal cell proliferation offers a unique interaction that can be manipulated to guide cells into a neuronal phenotype or to develop therapies for neuroblastomas.-Garwain, O., Valla, K., Scarlata, S. Phospholipase Cß1 regulates proliferation of neuronal cells.


G1 Phase , Gene Expression Regulation, Enzymologic , Neurites/enzymology , Phospholipase C beta/biosynthesis , S Phase , Animals , Cell Membrane/enzymology , Cell Membrane/genetics , Cell Membrane/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytosol/enzymology , Cytosol/pathology , Neurites/pathology , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/pathology , Neuroblastoma/therapy , PC12 Cells , Phospholipase C beta/genetics , Rats
7.
J Cell Physiol ; 233(1): 673-687, 2018 Jan.
Article En | MEDLINE | ID: mdl-28332716

The class IIa histone deacetylases (HDACs) play important roles in the central nervous system during diverse biological processes such as synaptic plasticity, axon regeneration, cell apoptosis, and neural differentiation. Although it is known that HDAC5 regulates neuronal differentiation, neither the physiological function nor the regulation of HDAC5 in neuronal differentiation is clear. Here, we identify HDAC5 as an inhibitor of neurite elongation and show that HDAC5 is regulated by the brain enriched microRNA miR-124 and miR-9. We discover that HDAC5 inhibits neurite extension both in differentiated P19 cells and primary neurons. We also show that the neuronal membrane glycoprotein GPM6A (M6a) is a direct target gene of HDAC5 regulated transcriptional factor MEF2C. HDAC5 inhibits neurite elongation, acting at least partially via a MEF2C/M6a signaling pathway. We also confirmed the miR-124/miR-9 regulated HDAC5-MEF2C-M6a pathway regulates neurite development in primary neurons. Thus, HDAC5 emerges as a cellular conductor of MEF2C and M6a activity and is regulated by miR-124 and miR-9 to control neurite development.


Embryonic Stem Cells/enzymology , Histone Deacetylases/metabolism , Membrane Glycoproteins/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/enzymology , Neurites/enzymology , Neurogenesis , Animals , Down-Regulation , Embryonic Stem Cells/drug effects , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gestational Age , HEK293 Cells , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/drug effects , Neurites/drug effects , Neurogenesis/drug effects , Signal Transduction , Transfection
8.
Mol Cell Biochem ; 444(1-2): 1-13, 2018 Jul.
Article En | MEDLINE | ID: mdl-29159770

The JNK-interacting protein 3 (JIP3) is a molecular scaffold, expressed predominantly in neurons, that serves to coordinate the activation of the c-Jun N-terminal kinase (JNK) by binding to JNK and the upstream kinases involved in its activation. The JNK pathway is involved in the regulation of many cellular processes including the control of cell survival, cell death and differentiation. JIP3 also associates with microtubule motor proteins such as kinesin and dynein and is likely an adapter protein involved in the tethering of vesicular cargoes to the motors involved in axonal transport in neurons. We have used immunofluorescence microscopy and biochemical fractionation to investigate the subcellular distribution of JIP3 in relation to JNK and to vesicular and organelle markers in rat pheochromocytoma cells (PC12) differentiating in response to nerve growth factor. In differentiated PC12 cells, JIP3 was seen to accumulate in growth cones at the tips of developing neurites where it co-localised with both JNK and the JNK substrate paxillin. Cellular fractionation of PC12 cells showed that JIP3 was associated with a subpopulation of vesicles in the microsomal fraction, distinct from synaptic vesicles, likely to be an anterograde-directed exocytic vesicle pool. In differentiated PC12 cells, JIP3 did not appear to associate with retrograde endosomal vesicles thought to be involved in signalling axonal injury. Together, these observations indicate that JIP3 may be involved in transporting vesicular cargoes to the growth cones of PC12 cells, possibly targeting JNK to its substrate paxillin, and thus facilitating neurite outgrowth.


Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Growth Cones/enzymology , Nerve Tissue Proteins/metabolism , Neurites/enzymology , Secretory Vesicles/enzymology , Synaptic Vesicles/enzymology , Animals , PC12 Cells , Rats
9.
Cell Rep ; 20(9): 2169-2183, 2017 Aug 29.
Article En | MEDLINE | ID: mdl-28854366

The initiating events that promote tau mislocalization and pathology in Alzheimer's disease (AD) are not well defined, partly because of the lack of endogenous models that recapitulate tau dysfunction. We exposed wild-type neurons to a neuroinflammatory trigger and examined the effect on endogenous tau. We found that tau re-localized and accumulated within pathological neuritic foci, or beads, comprised of mostly hypo-phosphorylated, acetylated, and oligomeric tau. These structures were detected in aged wild-type mice and were enhanced in response to neuroinflammation in vivo, highlighting a previously undescribed endogenous age-related tau pathology. Strikingly, deletion or inhibition of the cytoplasmic shuttling factor HDAC6 suppressed neuritic tau bead formation in neurons and mice. Using mass spectrometry-based profiling, we identified a single neuroinflammatory factor, the metalloproteinase MMP-9, as a mediator of neuritic tau beading. Thus, our study uncovers a link between neuroinflammation and neuritic tau beading as a potential early-stage pathogenic mechanism in AD.


Histone Deacetylase 6/metabolism , Neurites/enzymology , Neurites/pathology , tau Proteins/metabolism , Acetylation , Aging/pathology , Amyloid beta-Peptides/toxicity , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Histone Deacetylase 6/antagonists & inhibitors , Humans , Inflammation/pathology , Mass Spectrometry , Mice, Knockout , Phosphorylation , Protein Multimerization , Stress, Physiological
10.
Dev Biol ; 430(1): 41-51, 2017 10 01.
Article En | MEDLINE | ID: mdl-28844905

Arginylation is an emerging protein modification mediated by arginyltransferase ATE1, shown to regulate embryogenesis and actin cytoskeleton, however its functions in different physiological systems are not well understood. Here we analyzed the role of ATE1 in brain development and neuronal growth by producing a conditional mouse knockout with Ate1 deletion in the nervous system driven by Nestin promoter (Nes-Ate1 mice). These mice were weaker than wild type, resulting in low postnatal survival rates, and had abnormalities in the brain that suggested defects in neuronal migration. Cultured Ate1 knockout neurons showed a reduction in the neurite outgrowth and the levels of doublecortin and F-actin in the growth cones. In wild type, ATE1 prominently localized to the growth cones, in addition to the cell bodies. Examination of the Ate1 mRNA sequence reveals the existence of putative zipcode-binding sequences involved in mRNA targeting to the cell periphery and local translation at the growth cones. Fluorescence in situ hybridization showed that Ate1 mRNA localized to the tips of the growth cones, likely due to zipcode-mediated targeting, and this localization coincided with spots of localization of arginylated ß-actin, which disappeared in the presence of protein synthesis inhibitors. We propose that zipcode-mediated co-targeting of Ate1 and ß-actin mRNA leads to localized co-translational arginylation of ß-actin that drives the growth cone migration and neurite outgrowth.


Aminoacyltransferases/metabolism , Brain/growth & development , Brain/metabolism , Growth Cones/enzymology , Neurites/enzymology , Neuronal Outgrowth , Actins/metabolism , Animals , Arginine/metabolism , Brain/abnormalities , Brain/pathology , Cell Movement , Doublecortin Domain Proteins , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Models, Biological , Neuropeptides/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
J Biol Chem ; 292(24): 9906-9918, 2017 06 16.
Article En | MEDLINE | ID: mdl-28432124

The R7 regulator of G protein signaling family (R7-RGS) critically regulates nervous system development and function. Mice lacking all R7-RGS subtypes exhibit diverse neurological phenotypes, and humans bearing mutations in the retinal R7-RGS isoform RGS9-1 have vision deficits. Although each R7-RGS subtype forms heterotrimeric complexes with Gß5 and R7-RGS-binding protein (R7BP) that regulate G protein-coupled receptor signaling by accelerating deactivation of Gi/o α-subunits, several neurological phenotypes of R7-RGS knock-out mice are not readily explained by dysregulated Gi/o signaling. Accordingly, we used tandem affinity purification and LC-MS/MS to search for novel proteins that interact with R7-RGS heterotrimers in the mouse brain. Among several proteins detected, we focused on Gα13 because it had not been linked to R7-RGS complexes before. Split-luciferase complementation assays indicated that Gα13 in its active or inactive state interacts with R7-RGS heterotrimers containing any R7-RGS isoform. LARG (leukemia-associated Rho guanine nucleotide exchange factor (GEF)), PDZ-RhoGEF, and p115RhoGEF augmented interaction between activated Gα13 and R7-RGS heterotrimers, indicating that these effector RhoGEFs can engage Gα13·R7-RGS complexes. Because Gα13/R7-RGS interaction required R7BP, we analyzed phenotypes of neuronal cell lines expressing RGS7 and Gß5 with or without R7BP. We found that neurite retraction evoked by Gα12/13-dependent lysophosphatidic acid receptors was augmented in R7BP-expressing cells. R7BP expression blunted neurite formation evoked by serum starvation by signaling mechanisms involving Gα12/13 but not Gαi/o These findings provide the first evidence that R7-RGS heterotrimers interact with Gα13 to augment signaling pathways that regulate neurite morphogenesis. This mechanism expands the diversity of functions whereby R7-RGS complexes regulate critical aspects of nervous system development and function.


Brain/metabolism , Carrier Proteins/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurons/metabolism , RGS Proteins/metabolism , Amino Acid Substitution , Animals , Brain/cytology , Brain/enzymology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , GTP-Binding Protein alpha Subunits, G12-G13/chemistry , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurites/enzymology , Neurons/cytology , Neurons/enzymology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Multimerization , RGS Proteins/chemistry , RGS Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction
12.
J Cell Biochem ; 118(11): 3960-3967, 2017 11.
Article En | MEDLINE | ID: mdl-28398601

In our work, we used an in vitro culture model to investigate whether antidepressant imipramine (Ip) may protect bupivacaine (Bv)-induced neurotoxicity in mouse dorsal root ganglion (DRG). Adult mouse DRG was treated with 5 mM Bv in vitro to induce neurotoxicity. DRG was then pre-treated with Ip, prior to Bv, to examine its effects on protecting Bv-induced DRG apoptosis and neurite degeneration. Ip-induced dynamic changes in Trk receptors, including TrkA/B/C and phosphor (p-)TrkA/B/C, were examined by qPCR and Western blot. TrkA and TrkB were inhibited by siRNAs to further investigate their functional role in Ip- and Bv-treated DRG. Ip protected Bv-induced apoptosis and neurite loss in DRG. Ip did not alter TrkA/B/C expressions, whereas significantly augmented protein productions of p-TrkA and p-TrkB, but not p-TrkC. SiRNA-mediated TrkA or TrkB downregulation inhibited Trk receptors, and reduced p-TrkA and p-TrkB in DRG. TrkA or TrkB downregulation alone had no effect on Ip-induced protection in Bv-injured DRG. However, co-inhibition of TrkA and TrkB significantly ameliorated the protective effect of Ip on Bv-induced apoptosis and neurite loss in DRG. Imipramine protected bupivacaine-induced neurotoxicity in DRG, likely via the co-activation of TrkA and TrkB signaling pathways. J. Cell. Biochem. 118: 3960-3967, 2017. © 2017 Wiley Periodicals, Inc.


Antidepressive Agents/pharmacology , Bupivacaine/adverse effects , Ganglia, Spinal/metabolism , Imipramine/pharmacology , Neurites/enzymology , Neurotoxicity Syndromes/prevention & control , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Animals , Bupivacaine/pharmacology , Enzyme Activation/drug effects , Ganglia, Spinal/pathology , Mice , Neurites/pathology , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Signal Transduction/drug effects
13.
Biochim Biophys Acta Gene Regul Mech ; 1860(6): 685-694, 2017 Jun.
Article En | MEDLINE | ID: mdl-28274785

The differential expression of mRNAs containing tandem alternative 3' UTRs, achieved by mechanisms of alternative polyadenylation and post-transcriptional regulation, has been correlated with a variety of cellular states. In differentiated cells and brain tissues there is a general use of distal polyadenylation signals, originating mRNAs with longer 3' UTRs, in contrast with proliferating cells and other tissues such as testis, where most mRNAs contain shorter 3' UTRs. Although cell type and state are relevant in many biological processes, how these mechanisms occur in specific brain cell types is still poorly understood. Rac1 is a member of the Rho family of small GTPases with essential roles in multiple cellular processes, including cell differentiation and axonal growth. Here we used different brain cell types and tissues, including oligodendrocytes, microglia, astrocytes, cortical and hippocampal neurons, and optical nerve, to show that classical Rho GTPases express mRNAs with alternative 3' UTRs differently, by gene- and cell- specific mechanisms. In particular, we show that Rac1 originate mRNA isoforms with longer 3' UTRs specifically during neurite growth of cortical, but not hippocampal neurons. Furthermore, we demonstrate that the longest Rac1 3' UTR is necessary for driving the mRNA to the neurites, and also for neurite outgrowth in cortical neurons. Our results indicate that the expression of Rac1 longer 3' UTR is a gene and cell-type specific mechanism in the brain, with a new physiological function in cortical neuron differentiation.


3' Untranslated Regions/physiology , Cerebral Cortex/enzymology , Gene Expression Regulation, Enzymologic/physiology , Neurites/enzymology , rac1 GTP-Binding Protein/biosynthesis , Animals , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/cytology , Humans , Rats , Rats, Wistar , rac1 GTP-Binding Protein/genetics
14.
Cell Mol Neurobiol ; 37(3): 417-426, 2017 Apr.
Article En | MEDLINE | ID: mdl-27098315

Minocycline, a semi-synthetic second-generation derivative of tetracycline, has been reported to exert neuroprotective effects both in animal models and in clinic trials of neurological diseases. In the present study, we first investigated the protective effects of minocycline on oxygen-glucose deprivation and reoxygenation-induced impairment of neurite outgrowth and its potential mechanism in the neuronal cell line, PC12 cells. We found that minocycline significantly increased cell viability, promoted neurite outgrowth and enhanced the expression of growth-associated protein-43 (GAP-43) in PC12 cells exposed to oxygen-glucose deprivation/reoxygenation injury. In addition, immunoblots revealed that minocycline reversed the overexpression of phosphorylated myosin light chain (MLC) and the suppression of activated extracellular signal-regulated kinase 1/2 (ERK1/2) caused by oxygen-glucose deprivation/reoxygenation injury. Moreover, the minocycline-induced neurite outgrowth was significantly blocked by Calyculin A (1 nM), an inhibitor of myosin light chain phosphatase (MLCP), but not by an ERK1/2 inhibitor (U0126; 10 µM). These findings suggested that minocycline activated the MLCP/MLC signaling pathway in PC12 cells after oxygen-glucose deprivation/reoxygenation injury, which resulted in the promotion of neurite outgrowth.


Glucose/deficiency , Minocycline/pharmacology , Myosin Light Chains/metabolism , Myosin-Light-Chain Phosphatase/metabolism , Neurites/pathology , Oxygen/pharmacology , Animals , Cell Death/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GAP-43 Protein/metabolism , Neurites/drug effects , Neurites/enzymology , PC12 Cells , Phosphorylation/drug effects , Rats
15.
J Biol Chem ; 291(46): 23895-23905, 2016 Nov 11.
Article En | MEDLINE | ID: mdl-27655914

Protein tyrosine phosphatase MEG2 (PTP-MEG2) is a unique nonreceptor tyrosine phosphatase associated with transport vesicles, where it facilitates membrane trafficking by dephosphorylation of the N-ethylmaleimide-sensitive fusion factor. In this study, we identify the neurotrophin receptor TrkA as a novel cargo whose transport to the cell surface requires PTP-MEG2 activity. In addition, TrkA is also a novel substrate of PTP-MEG2, which dephosphorylates both Tyr-490 and Tyr-674/Tyr-675 of TrkA. As a result, overexpression of PTP-MEG2 down-regulates NGF/TrkA signaling and blocks neurite outgrowth and differentiation in PC12 cells and cortical neurons.


Neurites/enzymology , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Receptor, trkA/metabolism , Signal Transduction/physiology , Animals , Mice , PC12 Cells , Protein Transport/physiology , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Rats
16.
Brain Res ; 1648(Pt A): 193-201, 2016 10 01.
Article En | MEDLINE | ID: mdl-27423518

Diacylglycerol kinase (DGK) is a lipid-metabolizing enzyme that phosphorylates diacylglycerol to produce phosphatidic acid. Previously, we reported that the δ isozyme of DGK was abundantly expressed in the mouse brain. However, the functions of DGKδ in the brain are still unclear. Because conventional DGKδ-knockout (KO) mice die within 24h after birth, we have generated brain-specific conditional DGKδ-KO mice to circumvent the lethality. In the novel object recognition test, the number of contacts in the DGKδ-KO mice to novel and familiar objects was greatly increased compared to the control mice, indicating that the DGKδ-KO mice showed irrational contacts with objects such as compulsive checking. In the marble burying test, which is used for analyzing obsessive-compulsive disorder (OCD)-like phenotypes, the DGKδ-KO mice buried more marbles than the control mice. Additionally, these phenotypes were significantly alleviated by the administration of an OCD remedy, fluoxetine. These results indicate that the DGKδ-KO mice showed OCD-like behaviors. Moreover, the number of long axon/neurites increased in both DGKδ-KO primary cortical neurons and DGKδ-knockdown neuroblastoma Neuro-2a cells compared to control cells. Conversely, overexpression of DGKδ decreased the number of long axon/neurites of Neuro-2a cells. Taken together, these results strongly suggest that a deficiency of DGKδ induces OCD-like behavior through enhancing axon/neurite outgrowth.


Behavior, Animal/physiology , Brain/enzymology , Diacylglycerol Kinase/physiology , Obsessive-Compulsive Disorder/enzymology , Animals , Behavior, Animal/drug effects , Cell Line, Tumor , Diacylglycerol Kinase/genetics , Female , Fluoxetine/administration & dosage , Isoenzymes/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/enzymology , Phenotype , Recognition, Psychology/physiology , Selective Serotonin Reuptake Inhibitors/administration & dosage
17.
J Cell Biol ; 212(1): 91-111, 2016 Jan 04.
Article En | MEDLINE | ID: mdl-26728857

Rho guanosine triphosphatases (GTPases) control the cytoskeletal dynamics that power neurite outgrowth. This process consists of dynamic neurite initiation, elongation, retraction, and branching cycles that are likely to be regulated by specific spatiotemporal signaling networks, which cannot be resolved with static, steady-state assays. We present NeuriteTracker, a computer-vision approach to automatically segment and track neuronal morphodynamics in time-lapse datasets. Feature extraction then quantifies dynamic neurite outgrowth phenotypes. We identify a set of stereotypic neurite outgrowth morphodynamic behaviors in a cultured neuronal cell system. Systematic RNA interference perturbation of a Rho GTPase interactome consisting of 219 proteins reveals a limited set of morphodynamic phenotypes. As proof of concept, we show that loss of function of two distinct RhoA-specific GTPase-activating proteins (GAPs) leads to opposite neurite outgrowth phenotypes. Imaging of RhoA activation dynamics indicates that both GAPs regulate different spatiotemporal Rho GTPase pools, with distinct functions. Our results provide a starting point to dissect spatiotemporal Rho GTPase signaling networks that regulate neurite outgrowth.


Neurites/enzymology , Signal Transduction , Spatio-Temporal Analysis , rhoA GTP-Binding Protein/metabolism , Animals , Mice , Neurites/metabolism , Phenotype , Tumor Cells, Cultured
18.
Hum Mol Genet ; 25(1): 109-22, 2016 Jan 01.
Article En | MEDLINE | ID: mdl-26546614

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common known cause of inherited Parkinson's disease (PD), and LRRK2 is a risk factor for idiopathic PD. How LRRK2 function is regulated is not well understood. Recently, the highly conserved 14-3-3 proteins, which play a key role in many cellular functions including cell death, have been shown to interact with LRRK2. In this study, we investigated whether 14-3-3s can regulate mutant LRRK2-induced neurite shortening and kinase activity. In the presence of 14-3-3θ overexpression, neurite length of primary neurons from BAC transgenic G2019S-LRRK2 mice returned back to wild-type levels. Similarly, 14-3-3θ overexpression reversed neurite shortening in neuronal cultures from BAC transgenic R1441G-LRRK2 mice. Conversely, inhibition of 14-3-3s by the pan-14-3-3 inhibitor difopein or dominant-negative 14-3-3θ further reduced neurite length in G2019S-LRRK2 cultures. Since G2019S-LRRK2 toxicity is likely mediated through increased kinase activity, we examined 14-3-3θ's effects on LRRK2 kinase activity. 14-3-3θ overexpression reduced the kinase activity of G2019S-LRRK2, while difopein promoted the kinase activity of G2019S-LRRK2. The ability of 14-3-3θ to reduce LRRK2 kinase activity required direct binding of 14-3-3θ with LRRK2. The potentiation of neurite shortening by difopein in G2019S-LRRK2 neurons was reversed by LRRK2 kinase inhibitors. Taken together, we conclude that 14-3-3θ can regulate LRRK2 and reduce the toxicity of mutant LRRK2 through a reduction of kinase activity.


14-3-3 Proteins/physiology , Neurites/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Enlargement , Cells, Cultured , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neurites/enzymology , Neurons/cytology , Neurons/metabolism , Parkinson Disease/metabolism , Phosphorylation , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/genetics , Proteins/genetics , Serine/metabolism
19.
Nature ; 526(7573): 443-7, 2015 Oct 15.
Article En | MEDLINE | ID: mdl-26322584

Alzheimer disease (AD) is characterized by the accumulation of amyloid plaques, which are predominantly composed of amyloid-ß peptide. Two principal physiological pathways either prevent or promote amyloid-ß generation from its precursor, ß-amyloid precursor protein (APP), in a competitive manner. Although APP processing has been studied in great detail, unknown proteolytic events seem to hinder stoichiometric analyses of APP metabolism in vivo. Here we describe a new physiological APP processing pathway, which generates proteolytic fragments capable of inhibiting neuronal activity within the hippocampus. We identify higher molecular mass carboxy-terminal fragments (CTFs) of APP, termed CTF-η, in addition to the long-known CTF-α and CTF-ß fragments generated by the α- and ß-secretases ADAM10 (a disintegrin and metalloproteinase 10) and BACE1 (ß-site APP cleaving enzyme 1), respectively. CTF-η generation is mediated in part by membrane-bound matrix metalloproteinases such as MT5-MMP, referred to as η-secretase activity. η-Secretase cleavage occurs primarily at amino acids 504-505 of APP695, releasing a truncated ectodomain. After shedding of this ectodomain, CTF-η is further processed by ADAM10 and BACE1 to release long and short Aη peptides (termed Aη-α and Aη-ß). CTFs produced by η-secretase are enriched in dystrophic neurites in an AD mouse model and in human AD brains. Genetic and pharmacological inhibition of BACE1 activity results in robust accumulation of CTF-η and Aη-α. In mice treated with a potent BACE1 inhibitor, hippocampal long-term potentiation was reduced. Notably, when recombinant or synthetic Aη-α was applied on hippocampal slices ex vivo, long-term potentiation was lowered. Furthermore, in vivo single-cell two-photon calcium imaging showed that hippocampal neuronal activity was attenuated by Aη-α. These findings not only demonstrate a major functionally relevant APP processing pathway, but may also indicate potential translational relevance for therapeutic strategies targeting APP processing.


Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/metabolism , Hippocampus/cytology , Matrix Metalloproteinases, Membrane-Associated/metabolism , Neurons/physiology , Proteolysis , ADAM Proteins/metabolism , ADAM10 Protein , Alzheimer Disease/enzymology , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/cerebrospinal fluid , Amyloid Precursor Protein Secretases/deficiency , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Protein Precursor/cerebrospinal fluid , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/deficiency , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Calcium Signaling , Disease Models, Animal , Female , Hippocampus/enzymology , Hippocampus/physiology , Humans , In Vitro Techniques , Long-Term Potentiation , Male , Matrix Metalloproteinases, Membrane-Associated/deficiency , Membrane Proteins/metabolism , Mice , Molecular Weight , Neurites/enzymology , Neurites/metabolism , Neurons/enzymology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plaque, Amyloid , Protein Processing, Post-Translational , Single-Cell Analysis
20.
Cell Mol Neurobiol ; 35(7): 987-93, 2015 Oct.
Article En | MEDLINE | ID: mdl-25862329

Liraglutide is the glucagon-like peptide-1 (GLP-1) synthetic form which has been approved by the US Food and Drug Administration to be released onto the market. The metabolic benefits of incretin hormone as an anti-diabetic agent are widely recognized, but its potential extra-pancreatic effects of GLP-1 analog (liraglutide) in the central nerve system are less well known. To this purpose, we used immunofluorescence method to examine the effect of liraglutide on neurite outgrowth in primary cortical neuron culture by measuring neurite length and confirmed the promotion effect. Then, we investigated the potential mechanisms and found that liraglutide promoted neurite outgrowth in a dose-dependant manner, and this effect could be partially inhibited by MEK-ERK inhibitor U0126. Besides, liraglutide induced an increase of p-ERK/ERK expression, which could be blocked in the presence of U0126. Similarly, phosphorylated transcription factor (p-CREB) level shared the same trend with p-ERK/ERK ratio after liraglutide treatment. Collectively, our data illustrated that that liraglutide exerts neurotrophin-like activity partly via MEK-ERK pathway, which might offer a novel idea for treatment of axon-associated neurological diseases.


Cerebral Cortex/drug effects , Liraglutide/pharmacology , MAP Kinase Signaling System/drug effects , Neurites/drug effects , Animals , Cells, Cultured , Cerebral Cortex/enzymology , Dose-Response Relationship, Drug , MAP Kinase Signaling System/physiology , Mice , Mice, Inbred C57BL , Neurites/enzymology
...