27 research outputs found

    Dissecting the Daily Feeding Pattern: Peripheral CLOCK/CYCLE Generate the Feeding/Fasting Episodes and Neuronal Molecular Clocks Synchronize Them

    Get PDF
    A 24-h rhythm of feeding behavior, or synchronized feeding/fasting episodes during the day, is crucial for survival. Internal clocks and light input regulate rhythmic behaviors, but how they generate feeding rhythms is not fully understood. Here we aimed to dissect the molecular pathways that generate daily feeding patterns. By measuring the semidiurnal amount of food ingested by single flies, we demonstrate that the generation of feeding rhythms under light:dark conditions requires quasimodo (qsm) but not molecular clocks. Under constant darkness, rhythmic feeding patterns consist of two components: CLOCK (CLK) in digestive/metabolic tissues generating feeding/fasting episodes, and the molecular clock in neurons synchronizing them to subjective daytime. Although CLK is a part of the molecular clock, the generation of feeding/fasting episodes by CLK in metabolic tissues was independent of molecular clock machinery. Our results revealed novel functions of qsm and CLK in feeding rhythms in Drosophila

    Stabilization of Microtubule-Unbound Tau via Tau Phosphorylation at Ser262/356 by Par-1/MARK Contributes to Augmentation of AD-Related Phosphorylation and Aβ42-Induced Tau Toxicity.

    Get PDF
    Abnormal accumulation of the microtubule-interacting protein tau is associated with neurodegenerative diseases including Alzheimer\u27s disease (AD). β-amyloid (Aβ) lies upstream of abnormal tau behavior, including detachment from microtubules, phosphorylation at several disease-specific sites, and self-aggregation into toxic tau species in AD brains. To prevent the cascade of events leading to neurodegeneration in AD, it is essential to elucidate the mechanisms underlying the initial events of tau mismetabolism. Currently, however, these mechanisms remain unclear. In this study, using transgenic Drosophila co-expressing human tau and Aβ, we found that tau phosphorylation at AD-related Ser262/356 stabilized microtubule-unbound tau in the early phase of tau mismetabolism, leading to neurodegeneration. Aβ increased the level of tau detached from microtubules, independent of the phosphorylation status at GSK3-targeted SP/TP sites. Such mislocalized tau proteins, especially the less phosphorylated species, were stabilized by phosphorylation at Ser262/356 via PAR-1/MARK. Levels of Ser262 phosphorylation were increased by Aβ42, and blocking this stabilization of tau suppressed Aβ42-mediated augmentation of tau toxicity and an increase in the levels of tau phosphorylation at the SP/TP site Thr231, suggesting that this process may be involved in AD pathogenesis. In contrast to PAR-1/MARK, blocking tau phosphorylation at SP/TP sites by knockdown of Sgg/GSK3 did not reduce tau levels, suppress tau mislocalization to the cytosol, or diminish Aβ-mediated augmentation of tau toxicity. These results suggest that stabilization of microtubule-unbound tau by phosphorylation at Ser262/356 via the PAR-1/MARK may act in the initial steps of tau mismetabolism in AD pathogenesis, and that such tau species may represent a potential therapeutic target for AD

    Loss of axonal mitochondria promotes tau-mediated neurodegeneration and Alzheimer\u27s disease-related tau phosphorylation via PAR-1.

    Get PDF
    Abnormal phosphorylation and toxicity of a microtubule-associated protein tau are involved in the pathogenesis of Alzheimer\u27s disease (AD); however, what pathological conditions trigger tau abnormality in AD is not fully understood. A reduction in the number of mitochondria in the axon has been implicated in AD. In this study, we investigated whether and how loss of axonal mitochondria promotes tau phosphorylation and toxicity in vivo. Using transgenic Drosophila expressing human tau, we found that RNAi-mediated knockdown of milton or Miro, an adaptor protein essential for axonal transport of mitochondria, enhanced human tau-induced neurodegeneration. Tau phosphorylation at an AD-related site Ser262 increased with knockdown of milton or Miro; and partitioning defective-1 (PAR-1), the Drosophila homolog of mammalian microtubule affinity-regulating kinase, mediated this increase of tau phosphorylation. Tau phosphorylation at Ser262 has been reported to promote tau detachment from microtubules, and we found that the levels of microtubule-unbound free tau increased by milton knockdown. Blocking tau phosphorylation at Ser262 site by PAR-1 knockdown or by mutating the Ser262 site to unphosphorylatable alanine suppressed the enhancement of tau-induced neurodegeneration caused by milton knockdown. Furthermore, knockdown of milton or Miro increased the levels of active PAR-1. These results suggest that an increase in tau phosphorylation at Ser262 through PAR-1 contributes to tau-mediated neurodegeneration under a pathological condition in which axonal mitochondria is depleted. Intriguingly, we found that knockdown of milton or Miro alone caused late-onset neurodegeneration in the fly brain, and this neurodegeneration could be suppressed by knockdown of Drosophila tau or PAR-1. Our results suggest that loss of axonal mitochondria may play an important role in tau phosphorylation and toxicity in the pathogenesis of AD

    EDEM Function in ERAD Protects against Chronic ER Proteinopathy and Age-Related Physiological Decline in Drosophila

    No full text
    Summary The unfolded protein response (UPR), which protects cells against accumulation of misfolded proteins in the ER, is induced in several age-associated degenerative diseases. However, sustained UPR activation has negative effects on cellular functions and may worsen disease symptoms. It remains unknown whether and how UPR components can be utilized to counteract chronic ER proteinopathies. We found that promotion of ER-associated degradation (ERAD) through upregulation of ERAD-enhancing α-mannosidase-like proteins (EDEMs) protected against chronic ER proteinopathy without inducing toxicity in a Drosophila model. ERAD activity in the brain decreased with aging, and upregulation of EDEMs suppressed age-dependent behavioral decline and extended the lifespan without affecting the UPR gene expression network. Intriguingly, EDEM mannosidase activity was dispensable for these protective effects. Therefore, upregulation of EDEM function in the ERAD protects against ER proteinopathy in vivo and thus represents a potential therapeutic target for chronic diseases

    PAR-1 stabilizes less phosphorylated forms of tau (tau<sub>lower</sub>) through phosphorylation at Ser262 and Ser356.

    No full text
    <p>(A) PAR-1 knockdown reduces the levels of tau with more prominent effect on tau<sub>lower</sub>. Western blots of fly heads expressing tau (tau) or that co-expressing tau and PAR-1RNAi (tau+PAR-1RNAi) with anti-tau antibody. (B) PAR-1 overexpression increases the levels of tau with more prominent effect on tau<sub>lower</sub>. Western blots of fly heads expressing tau (tau) or that co-expressing tau and PAR-1 (tau+PAR-1OE) with anti-tau antibody. (C) PAR-1 knockdown does not affect S2Atau levels. Western blots of fly heads expressing S2Atau (S2A) or that co-expressing S2Atau and PAR-1RNAi (S2A+PAR-1RNAi) with anti-tau antibody. (D) PAR-1 overexpression does not affect S2Atau levels. PAR-1 knockdown does not affect S2Atau levels. Western blots of fly heads expressing S2Atau (S2A) or that co-expressing S2Atau and PAR-1 (S2A+PAR-1OE) with anti-tau antibody. Tubulin or actin was used as loading control. Mean ± SD, n = 4–5; *, <i>p</i> < 0.05, ***, <i>p</i> < 0.005, n.s., not significant (<i>p</i> > 0.05) by Student's t-test. Representative blots are shown. Transgene expression was driven by gmr-GAL4.</p

    Blocking tau phosphorylation at Ser262/356 with unphosphorylatable alanine substitutions (S2A) preferentially reduces the levels of tau<sub>lower</sub> and partially suppresses the Aβ42-mediated increase in tau mislocalization from the microtubule to the cytosol.

    No full text
    <p>(A) Expression of either S2Atau (S2A) alone or co-expression of S2Atau and Aβ42 (S2A+Aβ42) does not cause eye degeneration. No significant difference in the surface area of the external eyes between S2A and S2A+ Aβ42 (mean ± SE, n = 6–8, N.S., not significant (<i>p</i> > 0.05) by one-way ANOVA). Transgene expression was driven by gmr-GAL4. (B) S2Atau shows different phosphorylation profiles compared to wild-type tau. Wild-type tau or S2Atau were co-expressed with Aβ42 (tau+Aβ42 and Aβ42+S2A, respectively) and subjected to western blotting with pan-tau antibody (tauC and tau46) or antibodies that recognize phosphorylation status of tau at the specific sites (pSer262, TAU-1 and pSer202). (C) The ratio of signal intensities of tau<sub>lower</sub> to tau<sup>upper</sup> detected by tauC. (D) The ratio of signal intensities of TAU-1 blot to tauC blot. Mean ± SD, n = 5; ***, <i>p</i> < 0.005 by Student's t-test. (E) The ratio of signal intensities of pSer202 blot to tauC blot. Mean ± SD, n = 5; ***, <i>p</i> < 0.005 by Student's t-test. (F) Aβ42 increased the level of S2Atau in the cytosol and decreased those in the microtubule fraction, while the effects were smaller than those in wild-type tau. Mean ± SD, n = 4; *, <i>p</i> < 0.05, ***, <i>p</i> < 0.005 compared by Student's t-test. Representative blots are shown. Transgene expression was driven by gmr-GAL4.</p
    corecore