31 research outputs found

    The Microtubule Regulatory Protein Stathmin Is Required to Maintain the Integrity of Axonal Microtubules in <i>Drosophila</i>

    Get PDF
    <div><p>Axonal transport, a form of long-distance, bi-directional intracellular transport that occurs between the cell body and synaptic terminal, is critical in maintaining the function and viability of neurons. We have identified a requirement for the stathmin (<i>stai</i>) gene in the maintenance of axonal microtubules and regulation of axonal transport in <i>Drosophila</i>. The <i>stai</i> gene encodes a cytosolic phosphoprotein that regulates microtubule dynamics by partitioning tubulin dimers between pools of soluble tubulin and polymerized microtubules, and by directly binding to microtubules and promoting depolymerization. Analysis of <i>stai</i> function in <i>Drosophila</i>, which has a single <i>stai</i> gene, circumvents potential complications with studies performed in vertebrate systems in which mutant phenotypes may be compensated by genetic redundancy of other members of the <i>stai</i> gene family. This has allowed us to identify an essential function for <i>stai</i> in the maintenance of the integrity of axonal microtubules. In addition to the severe disruption in the abundance and architecture of microtubules in the axons of <i>stai</i> mutant <i>Drosophila</i>, we also observe additional neurological phenotypes associated with loss of <i>stai</i> function including a posterior paralysis and tail-flip phenotype in third instar larvae, aberrant accumulation of transported membranous organelles in <i>stai</i> deficient axons, a progressive bang-sensitive response to mechanical stimulation reminiscent of the class of <i>Drosophila</i> mutants used to model human epileptic seizures, and a reduced adult lifespan. Reductions in the levels of Kinesin-1, the primary anterograde motor in axonal transport, enhance these phenotypes. Collectively, our results indicate that <i>stai</i> has an important role in neuronal function, likely through the maintenance of microtubule integrity in the axons of nerves of the peripheral nervous system necessary to support and sustain long-distance axonal transport.</p> </div

    Loss of <i>stai</i> Function Results in Defects in Fast Axonal Transport.

    No full text
    <p>Mutations in <i>stai</i> result in phenotypes consistent with axonal transport defects. (<b>A</b>) Wildtype third instar larvae maintain a flat body posture when crawling. In contrast, homozygous <i>stai</i><sup><i>B200</i></sup> larvae (<b>B</b>) and <i>stai</i><sup><i>rdtp</i></sup> larvae (<b>C</b>) exhibit a posterior paralysis “tail flip” phenotype common for <i>Drosophila</i> mutants defective in axonal transport (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0068324#pone.0068324.s002" target="_blank">Movies S1</a>-S3). The tail-flip phenotype is also observed in heterozygous <i>stai</i><sup><i>B200</i></sup>/<i>stai</i><sup><i>rdtp</i></sup> larvae (<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0068324#pone.0068324.s005" target="_blank">Movie S4</a>). Confocal micrographs of sections of segmental nerve from wildtype (<b>D</b>), <i>stai</i><sup><i>B200</i></sup><i>/+</i> heterozygous (<b>E</b>) and <i>stai</i><sup><i>B200</i></sup> homozygous (<b>F</b>) third instar larvae stained with antibody against synaptic vesicle protein cysteine string protein (CSP). (<b>D</b>) The axons of wild type animals showed faint, punctate staining for CSP uniformly throughout the axons within the compound nerve bundle. (<b>E</b>) Although heterozygous <i>stai</i><sup><i>B200</i></sup> larvae showed no obvious sluggishness or tail-flip phenotype when they crawled, mild accumulations of CSP were observed in some segmental nerve axons. (<b>F</b>) In contrast, homozygous mutant <i>stai</i><sup><i>B200</i></sup> larvae exhibit abundant accumulations of CSP throughout the segmental nerve, confirming a disruption in axonal transport. (<b>G</b>) When <i>stai</i><sup><i>B200</i></sup> is placed in heterozygous combination with <i>Df</i>(<i>2L</i>) <i>Exel6015</i>, a chromosomal deletion that spans the <i>stai</i> locus, there is an enhancement in the abundance of axonal accumulation of CSP. (<b>H</b>) The severity of axonal transport defects was quantified by averaging the number of accumulations of CSP immunopositive anterograde and retrograde membranous axonal cargos observed in the segmental nerve axons of <i>stai</i> deficient third instar larvae. To determine if there was an anterior-to-posterior gradient in the severity of axonal transport defects, regions of segmental nerve axons were analyzed as they passed through abdominal segment A2 (light grey bars) and abdominal segment A4 (dark grey bars). Data is also presented as total number of accumulations observed in both abdominal regions (black bars). The only significant regional difference observed in the abundance of axonal clogs in segmental nerve axons between abdominal segment A2 and A4 was in the axons examined from <i>stai</i><sup><i>B200</i></sup><i>/Df</i>(<i>2L</i>) <i>Exel6015 larvae</i> (<i>p<0.0001</i>). Wild type larvae exhibited 0.57 ± 1.21 axonal clogs/50 µm segmental nerve axon in all axons examined. Homozygous <i>stai</i><sup><i>B200</i></sup> larvae averaged 13.55 ± 5.56 axonal clogs/50 µm segmental nerve axon (p<0.0001). The average number of axonal clogs observed in the segmental nerve axons of <i>stai</i><sup><i>B200</i></sup><i>/Df</i>(<i>2L</i>) <i>Exel6015</i> larvae was 15.10 ± 6.07 axonal clogs/50 µm segmental nerve axon. In panels A-C, the scale bar = 1mm. In panels D-G, the scale bar = 10 µm.</p

    Loss of <i>stai</i> Function Reduces Levels of Stabilized Axonal Microtubules.

    No full text
    <p>(<b>A</b>–<b>F</b>) Confocal micrographs of segmental nerve axons from third instar larvae immunostained with antibody against acetylated α-tubulin (mAb 611B1). Axons from (<b>A</b>) wildtype, (<b>B</b>) <i>stai</i><sup><i>B200</i></sup> and (<b>C</b>) <i>stai</i><sup><i>B200</i></sup><i>/Df</i>(<i>2L</i>) <i>Exel6015</i> larvae. (<b>D</b>) Precise excision of the <i>piggyBac</i> element <i>PBac</i>{<i>5HPw<sup>+</sup></i>}<sup><i>B200</i></sup> reverts the reduction in acetylated α-tubulin back to wild type levels. The reduction in acetylated α-tubulin staining intensity observed in <i>stai</i> deficient segmental nerve axons is significantly restored with the ubiquitous ectopic expression of <i>Drosophila staiB2</i> (<b>E</b>) or human STMN1 (<b>F</b>). (<b>G</b>) Quantification of average pixel intensity of acetylated α-tubulin immunostained axons of third instar larval segmental nerves. Results are presented in arbitrary units (a.u.) of fluorescence intensity (mean ± S.D.). In panels A-F the scale bar = 10 µm.</p

    Identification of Mutations in the <i>stathmin</i> (<i>stai</i>) Locus.

    No full text
    <p>(<b>A</b>) Genomic structure of the <i>Drosophila</i><i>stai</i> locus and the positions of the mutagenic copia retrotransposon <i>stai</i><sup><i>rdtp</i></sup> and the <i>piggyBac</i> element <i>stai</i><sup><i>B200</i></sup>. Exons are boxed, noncoding portions of exons are white, exons 3, 4, 5, shared by all stai proteins, are light grey (after Lachkar et al, 2010). Transcripts encoding <i>staiA</i> isoforms include exons 1 and 2 with alternative splicing that either includes (<i>staiA1</i>) or excludes (<i>staiA2</i>) exon 6. Transcripts encoding <i>staiB</i> isoforms include exons 1’and 2’ with alternative splicing that either includes (<i>staiB1</i>) or excludes (<i>staiB2</i>) exon 6. The copia retrotransposon <i>stai<sup>rdtp</sup></i> is inserted in the open reading frame of the <i>stai</i> gene in exon 1', ten base pairs downstream of the translational start site used to produce nervous system enriched <i>staiB</i> encoding transcripts. The <i>piggyBac</i> element <i>PBac</i>{<i>5HPw<sup>+</sup></i>}<i>stai<sup>B200</sup></i> is inserted in the 2.8 kb intron separating exons 1’ and 2’, 1.3kb downstream of the splice junction of exon 1’. (<b>B</b>) The copia insertion in the <i>stai</i> gene was identified by PCR amplification across the open reading frame of <i>stai</i> exon 1’ that resulted in an unexpectedly large 5.6 kb product from genomic DNA isolated from <i>stai</i><sup><i>rdtp</i></sup> homozygous larvae. The black arrowheads in Figure 1A represent the relative position of PCR primers used. (<b>C</b>) qRT-PCR of <i>staiA</i> and <i>staiB</i> mRNA derived from third instar larvae is shown. The expression of <i>staiA</i> and <i>staiB</i> is significantly reduced in all <i>stai</i> mutant genotypes analyzed compared to wild type expression levels (P<0.01). The red and blue arrowheads in Figure 1A represent the relative position of primers used for qRT-PCR of <i>staiA</i> and <i>staiB</i> message. Results are normalized against the expression of the <i>GAPDH</i> gene product and are presented as mean ± SD.</p

    Loss of <i>stai</i> Function Reduces Futsch (MAP1B) Protein Levels in Axons and Alters the Integrity of Axonal Microtubules.

    No full text
    <p>(<b>A</b>–<b>F</b>) Confocal micrographs of segmental nerve axons from third instar larvae immunostained with antibody against the MAP Futsch (mAb 22c10). (<b>A</b>) Wild type larvae have well defined Futsch staining in individual axons within segmental nerve bundles. (<b>B</b>) In contrast, loss of <i>stai</i> function results in an inconsistent distribution of Futsch protein along the length of individual axons within segmental nerve bundles. In some regions of an axon, there is prominent Futsch staining (arrows) whereas Futsch staining is noticeably reduced in other areas of the same axon (small arrowheads). In addition, loss of <i>stai</i> function causes gross perturbations in axon morphology (large arrowheads). (<b>C</b>) The perturbations in axon morphology are more profound in segmental nerve axons from <i>stai</i><sup><i>B200</i></sup><i>/Df</i>(<i>2L</i>) <i>Exel6015</i> larvae. (<b>D</b>) Precise excision of the <i>piggyBac</i> element <i>PBac</i>{<i>5HPw<sup>+</sup></i>}<sup><i>B200</i></sup> reverts the observed phenotypes to wildtype. The reduction in Futsch staining intensity and perturbations in axon morphology observed in <i>stai</i> deficient segmental nerve axons can be ameliorated with the ubiquitous ectopic expression of <i>Drosophila staiB2</i> (<b>E</b>) or human STMN1 (<b>F</b>). (<b>G</b>) Quantification of average pixel intensity of Futsch immunostained axons of third instar larval segmental nerves. Results are presented in arbitrary units (a.u.) of fluorescence intensity (mean ± S.D.). In panels A-F the scale bar = 10 µm.</p

    Glycine transporter 1 is a target for the treatment of epilepsy

    No full text
    Glycine is the major inhibitory neurotransmitter in brainstem and spinal cord, whereas in hippocampus glycine exerts dual modulatory roles on strychnine-sensitive glycine receptors and on the strychnine-insensitive glycineB site of the N-methyl-D-aspartate receptor (NMDAR). In hippocampus, the synaptic availability of glycine is largely under control of glycine transporter 1 (GlyT1). Since epilepsy is a disorder of disrupted network homeostasis affecting the equilibrium of various neurotransmitters and neuromodulators, we hypothesized that changes in hippocampal GlyT1 expression and resulting disruption of glycine homeostasis might be implicated in the pathophysiology of epilepsy. Using two different rodent models of temporal lobe epilepsy (TLE)--the intrahippocampal kainic acid model of TLE in mice, and the rat model of tetanic stimulation-induced TLE--we first demonstrated robust overexpression of GlyT1 in the hippocampal formation, suggesting dysfunctional glycine signaling in epilepsy. Overexpression of GlyT1 in the hippocampal formation was corroborated in human TLE samples by quantitative real time PCR. In support of a role of dysfunctional glycine signaling in the pathophysiology of epilepsy, both the genetic deletion of GlyT1 in hippocampus and the GlyT1 inhibitor LY2365109 increased seizure thresholds in mice. Importantly, chronic seizures in the mouse model of TLE were robustly suppressed by systemic administration of the GlyT1 inhibitor LY2365109. We conclude that GlyT1 overexpression in the epileptic brain constitutes a new target for therapeutic intervention, and that GlyT1 inhibitors constitute a new class of antiictogenic drugs. These findings are of translational value since GlyT1 inhibitors are already in clinical development to treat cognitive symptoms in schizophreni
    corecore