60 research outputs found

    Developmental expression of COE across the Metazoa supports a conserved role in neuronal cell-type specification and mesodermal development

    Get PDF
    The transcription factor COE (collier/olfactory-1/early B cell factor) is an unusual basic helixā€“loopā€“helix transcription factor as it lacks a basic domain and is maintained as a single copy gene in the genomes of all currently analysed non-vertebrate Metazoan genomes. Given the unique features of the COE gene, its proposed ancestral role in the specification of chemosensory neurons and the wealth of functional data from vertebrates and Drosophila, the evolutionary history of the COE gene can be readily investigated. We have examined the ways in which COE expression has diversified among the Metazoa by analysing its expression from representatives of four disparate invertebrate phyla: Ctenophora (Mnemiopsis leidyi); Mollusca (Haliotis asinina); Annelida (Capitella teleta and Chaetopterus) and Echinodermata (Strongylocentrotus purpuratus). In addition, we have studied COE function with knockdown experiments in S. purpuratus, which indicate that COE is likely to be involved in repressing serotonergic cell fate in the apical ganglion of dipleurula larvae. These analyses suggest that COE has played an important role in the evolution of ectodermally derived tissues (likely primarily nervous tissues) and mesodermally derived tissues. Our results provide a broad evolutionary foundation from which further studies aimed at the functional characterisation and evolution of COE can be investigated

    Endogenous anabolic hormone responses to endurance versus resistance exercise and training in women

    No full text
    Genome-wide association studies implicate the MIR137HG risk variant rs1625579 (MIR137HGrv) within the host gene for microRNA-137 as a potential regulator of schizophrenia susceptibility. We examined the influence of MIR137HGrv genotype on 17 subcortical and callosal volumes in a large sample of individuals with schizophrenia and healthy controls (n=841). Although the volumes were overall reduced relative to healthy controls, for individuals with schizophrenia the homozygous MIR137HGrv risk genotype was associated with attenuated reduction of mid-posterior corpus callosum volume (p=0.001), along with trend-level effects in the adjacent central and posterior corpus callosum. These findings are unique in the literature and remain robust after analysis in ethnically homogenous and single-scanner subsets of the larger sample. Thus, our study suggests that the mechanisms whereby MIR137HGrv works to increase schizophrenia risk are not those that generate the corpus callosum volume reductions commonly found in the disorder

    PKCĪµ Promotes HuD-Mediated Neprilysin mRNA Stability and Enhances Neprilysin-Induced AĪ² Degradation in Brain Neurons

    Get PDF
    Amyloid-beta (AĪ²) peptide accumulation in the brain is a pathological hallmark of all forms of Alzheimer's disease. An imbalance between AĪ² production and clearance from the brain may contribute to accumulation of neurotoxic AĪ² and subsequent synaptic loss, which is the strongest correlate of the extent of memory loss in AD. The activity of neprilysin (NEP), a potent AĪ²-degrading enzyme, is decreased in the AD brain. Expression of HuD, an mRNA-binding protein important for synaptogenesis and neuronal plasticity, is also decreased in the AD brain. HuD is regulated by protein kinase CĪµ (PKCĪµ), and we previously demonstrated that PKCĪµ activation decreases AĪ² levels. We hypothesized that PKCĪµ acts through HuD to stabilize NEP mRNA, modulate its localization, and support NEP activity. Conversely, loss of PKCĪµ-activated HuD in AD leads to decreased NEP activity and accumulation of AĪ². Here we show that HuD is associated with NEP mRNA in cultures of human SK-N-SH cells. Treatment with bryostatin, a PKCĪµ-selective activator, enhanced NEP association with HuD and increased NEP mRNA stability. Activation of PKCĪµ also increased NEP protein levels, increased NEP phosphorylation, and induced cell surface expression. In addition, specific PKCĪµ activation directly stimulated NEP activity, leading to degradation of a monomeric form of AĪ² peptide and decreased AĪ² neuronal toxicity, as measured by cell viability. Bryostatin treatment also rescued AĪ²-mediated inhibition of HuD-NEP mRNA binding, NEP protein expression, and NEP cell membrane translocation. These results suggest that PKCĪµ activation reduces AĪ² by up-regulating, via the mRNA-binding protein HuD, AĪ²-degrading enzymes such as NEP. Thus, PKCĪµ activation may have therapeutic efficacy for AD by reducing neurotoxic AĪ² accumulation as well as having direct anti-apoptotic and synaptogenic effects
    • ā€¦
    corecore