Elsevier

Neurobiology of Disease

Volume 77, May 2015, Pages 117-126
Neurobiology of Disease

Manipulations of amyloid precursor protein cleavage disrupt the circadian clock in aging Drosophila

https://doi.org/10.1016/j.nbd.2015.02.012Get rights and content

Highlights

  • dBACE overexpression accelerates decline of rest-activity rhythms in aging Drosophila.

  • PER oscillations in pacemaker neurons are dampened by elevated dBACE.

  • Kuzbanian overexpression induces age-related rest-activity rhythm decline in flies.

  • Neuronal dAICD expression induces age-related rest-activity rhythm decline.

  • APPL expression in pacemaker neurons strengthens rest-activity rhythms in aging flies.

Abstract

Alzheimer's disease (AD) is a neurodegenerative disease characterized by severe cognitive deterioration. While causes of AD pathology are debated, a large body of evidence suggests that increased cleavage of Amyloid Precursor Protein (APP) producing the neurotoxic Amyloid-β (Aβ) peptide plays a fundamental role in AD pathogenesis. One of the detrimental behavioral symptoms commonly associated with AD is the fragmentation of sleep-activity cycles with increased nighttime activity and daytime naps in humans. Sleep-activity cycles, as well as physiological and cellular rhythms, which may be important for neuronal homeostasis, are generated by a molecular system known as the circadian clock. Links between AD and the circadian system are increasingly evident but not well understood. Here we examined whether genetic manipulations of APP-like (APPL) protein cleavage in Drosophila melanogaster affect rest-activity rhythms and core circadian clock function in this model organism. We show that the increased β-cleavage of endogenous APPL by the β-secretase (dBACE) severely disrupts circadian behavior and leads to reduced expression of clock protein PER in central clock neurons of aging flies. Our data suggest that behavioral rhythm disruption is not a product of APPL-derived production but rather may be caused by a mechanism common to both α and β-cleavage pathways. Specifically, we show that increased production of the endogenous Drosophila Amyloid Intracellular Domain (dAICD) caused disruption of circadian rest-activity rhythms, while flies overexpressing endogenous APPL maintained stronger circadian rhythms during aging. In summary, our study offers a novel entry point toward understanding the mechanism of circadian rhythm disruption in Alzheimer's disease.

Introduction

Alzheimer's disease (AD) is characterized by progressive neurodegeneration resulting in the loss of cognitive ability. The exact pathology of AD is not well understood and is widely debated. The amyloid cascade hypothesis suggests that abnormal production of neurotoxic Amyloid-beta (Aβ) in combination with tangles of phosphorylated Tau-microtubule associated protein lead to neuronal dysfunction, cell loss and thus cognitive decline (Hardy and Selkoe, 2002, Mandelkow and Mandelkow, 1998, Selkoe, 2000). The crucial enzyme in the production of Aβ is the rate-limiting Beta-site Amyloid Precursor Protein Cleaving Enzyme (BACE), which shows elevated expression in AD patients (Fukumoto et al., 2002, Vassar et al., 2009). Recently, knock-in of human BACE was shown to recapitulate many behavioral and physiological phenotypes of AD in a murine model (Plucinska et al., 2014). In addition to BACE, Amyloid Precursor Protein (APP) is cleaved by an α-secretase. Both cleavage pathways yield a secreted N-terminal fragment known as sAPP, sAPPβ for β-cleavage and sAPPα for α-cleavage. Subsequent ϒ-cleavage following β or α-cleavage results in either the Aβ or P3 fragment, respectively (De Strooper and Annaert, 2000, Selkoe, 2000, Turner et al., 2003). In addition to those two fragments, ϒ-cleavage also produces the APP intracellular domain (AICD), which has been shown to affect transcriptional regulation (Belyaev et al., 2010, Kimberly et al., 2001, Pardossi-Piquard and Checler, 2012, Turner et al., 2003). Both detrimental and positive effects of the AICD have been reported in cultured cells (Lu et al., 2000, Zhou et al., 2012); however, the physiological function of the AICD remains poorly understood.

One of the detrimental behavioral symptoms commonly associated with AD is the fragmentation of sleep-activity cycles with increased nighttime activity and daytime naps (Harper et al., 2005, Volicer et al., 2001, Wu and Swaab, 2007). Impaired rest-activity rhythms have also been reported in experimental AD model mice (Roh et al., 2012, Sterniczuk et al., 2010). Rest-activity cycles are generated by a molecular system known as the circadian clock. The circadian clock mechanism is based on negative feedback loops involving transcriptional activators and repressors, which are largely conserved from Drosophila to humans (Hardin and Panda, 2013). Circadian clocks generate daily rhythms in expression of many output genes leading to cellular, physiological, and behavioral rhythms. Loss of circadian rhythms is detrimental to health and adversely affects neuronal homeostasis in both murine (Hastings and Goedert, 2013, Kondratova and Kondratov, 2012, Reddy and O'Neill, 2010) and Drosophila models (Krishnan et al., 2012). Therefore, it is important to understand the connections between the circadian system and AD pathology.

The links between AD and circadian rhythms were inferred from transgenic model animals expressing pathogenic versions of human genes (Rezaval et al., 2008, Roh et al., 2012, Sterniczuk et al., 2010). Recent reports showed that pathogenic human amyloid peptides disrupt behavioral rest-activity rhythms in Drosophila but are not sufficient to disrupt molecular oscillations of PERIOD (PER) in central pacemaker neurons (Chen et al., 2014, Long et al., 2014). Here, we manipulated AD-related genes in the fruit fly, Drosophila melanogaster in order to understand endogenous pathways that may interfere with the circadian system. APP has a functional ortholog in Drosophila called Amyloid Precursor Protein-like (APPL) (Luo et al., 1990, Luo et al., 1992, Rosen et al., 1989). In addition, orthologs of BACE, the α-secretase, and the ϒ-secretase complexes have been identified in Drosophila. They are known as dBACE, Kuzbanian (KUZ) and Presenilin (PSN), respectively, and were shown to process APPL resulting in peptide fragments comparable to APP fragments (Bolkan et al., 2012, Carmine-Simmen et al., 2009, Greeve et al., 2004). This conservation made Drosophila a suitable model for our study. Because BACE is the rate-limiting enzyme in the production of Aβ, we first investigated if over-expression of dBACE is involved in the rest-activity disturbances characteristic of AD.

We found that increasing dBACE expression disrupted endogenous rest-activity rhythms. This effect was most severe in aged flies suggesting an age-dependent mechanism. Furthermore, dBACE expression resulted in the dampened oscillation of the core clock protein PER in central pacemaker neurons, which are master regulators of rest-activity rhythms. Surprisingly, over-expression of Kuzbanian (KUZ) also disrupted rest-activity rhythms suggesting a mechanism independent of endogenous Drosophila Aβ peptide (dAβ) production, which like its vertebrate ortholog has been shown to be neurotoxic (Carmine-Simmen et al., 2009). This suggested that rhythm deficits were due to a cleavage product of APPL conserved in both α and β cleavage pathways. Therefore, we expressed the APPL intracellular domain (dAICD) and this caused severe disruption of behavioral rest-activity rhythms. In contrast, expression of full-length APPL in central clock neurons protected against an age-dependent decline in rest-activity rhythms. Taken together, these findings suggest that APPL and specifically the dAICD can affect circadian behavior by interfering with molecular clock oscillations.

Section snippets

Fly stocks

D. melanogaster were reared on diet containing 1% agar, 6.25% cornmeal, 6.25% molasses, and 3.5% Red Star yeast at 25 °C. Flies were entrained to 12-hour light:dark (LD, 12:12) cycles (with an average light intensity of ~ 1500 lx). All experiments were performed on mated male flies of different ages, as specified in the results. We used the binary UAS–GAL4 system to express specific genes using transgenic flies carrying the following constructs: UAS–dBACE, UAS–APPL, UAS–dAICD (Carmine-Simmen et

Over-expression of dBACE in clock cells accelerates aging phenotypes and disrupts rest-activity rhythms

To examine effects of elevated dBACE in flies, we expressed dBACE in all clock cells using the timeless (tim-GAL4) driver. We first tested whether this affected lifespan, and found a significant reduction (p < 0.0001) in tim > dBACE flies with a median lifespan of 61d compared to 82d for tim > GFP controls (Fig. 1A). Aging also was accelerated in tim > dBACE flies as they had significantly reduced climbing ability (p < 0.0001) at 35d and 50d compared to age-matched controls (Fig. 1B). We next

Discussion

Loss of rest-activity rhythms is a well-established early symptom of AD in humans. Because disruption of circadian rhythms is detrimental to neuronal homeostasis (Kondratova and Kondratov, 2012, Krishnan et al., 2012), it is important to understand relationships between AD and circadian rhythms at the cellular and molecular levels. To address this question, we examined how manipulations of the fly ortholog of APP and its cleaving enzymes affect endogenous rest-activity rhythms and clock

Acknowledgments

We thank Dr. Taishi Yoshii for advice on immunocytochemistry, Dr. R Stanewsky for anti-PER antibody, Dr. Philip Copenhaver for anti-AICD antibody, and Dani Long for reading the manuscript. Research reported in this publication was supported by the National Institute of Aging of the National Institutes of Health under award number R01 AG045830 to JMG and by a pilot project grant from the Oregon Institute of Occupational Health Sciences to DK.

References (53)

  • E.M. Mandelkow et al.

    Tau in Alzheimer's disease

    Trends Cell Biol.

    (1998)
  • S. Martinek

    A role for the segment polarity gene shaggy/GSK-3 in the Drosophila circadian clock

    Cell

    (2001)
  • A.B. Reddy et al.

    Healthy clocks, healthy body, healthy mind

    Trends Cell Biol.

    (2010)
  • S.C. Renn

    A pdf neuropeptide gene mutation and ablation of PDF neurons each cause severe abnormalities of behavioral circadian rhythms in Drosophila [published erratum appears in Cell 2000 Mar 31;101(1): following 113]

    Cell

    (1999)
  • R. Sterniczuk

    Characterization of the 3xTg-AD mouse model of Alzheimer's disease: Part 1. Circadian changes

    Brain Res.

    (2010)
  • P.R. Turner

    Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory

    Prog. Neurobiol.

    (2003)
  • J.S. Wentzell

    Amyloid precursor proteins are protective in Drosophila models of progressive neurodegeneration

    Neurobiol. Dis.

    (2012)
  • Y.H. Wu et al.

    Disturbance and strategies for reactivation of the circadian rhythm system in aging and Alzheimer's disease

    Sleep Med.

    (2007)
  • F. Zhou

    The APP intracellular domain (AICD) inhibits Wnt signalling and promotes neurite outgrowth

    Biochim. Biophys. Acta

    (2012)
  • B.J. Bolkan

    Beta-secretase cleavage of the fly amyloid precursor protein is required for glial survival

    J. Neurosci.

    (2012)
  • K.F. Chen

    The central molecular clock is robust in the face of behavioural arrhythmia in a Drosophila model of Alzheimer's disease

    Dis. Model. Mech.

    (2014)
  • B. De Strooper et al.

    Proteolytic processing and cell biological functions of the amyloid precursor protein

    J. Cell Sci.

    (2000)
  • R. Epis

    Alpha, beta- and gamma-secretases in Alzheimer's disease

    Front. Biosci. (Schol. Ed.)

    (2012)
  • H. Fukumoto

    Beta-secretase protein and activity are increased in the neocortex in Alzheimer disease

    Arch. Neurol.

    (2002)
  • K. Ghosal

    APP intracellular domain impairs adult neurogenesis in transgenic mice by inducing neuroinflammation

    PLoS One

    (2010)
  • V. Goguel

    Drosophila amyloid precursor protein-like is required for long-term memory

    J. Neurosci.

    (2011)
  • Cited by (26)

    • Animal Models of Alzheimer's Disease

      2017, Animal Models for the Study of Human Disease: Second Edition
    • Candidate mechanisms underlying the association between sleep-wake disruptions and Alzheimer's disease

      2017, Sleep Medicine Reviews
      Citation Excerpt :

      The mice also exhibited altered SCN expression patterns of the circadian clock genes, e.g., Bmal1, which through in vitro tests was corroborated to result from Aβ-induced degradation [106]. Similar findings have also been observed following overexpression of orthologs of APP cleavage enzymes in drosophila flies, in which the resulting increase in Aβ production resulted in disrupted circadian rhythms [107]. Collectively, evidence from both human and animal studies provide a strong rationale for hypothesizing that poor sleep and disrupted circadian rhythms may be a potential early marker of neuropathology during the long preclinical phase of AD.

    • Sleep and Memory Formation in Drosophila

      2017, Learning and Memory: A Comprehensive Reference
    • Enhanced sleep reverses memory deficits and underlying pathology in drosophila models of Alzheimer's disease

      2017, Neurobiology of Sleep and Circadian Rhythms
      Citation Excerpt :

      All statistically different groups are defined as *P<0.05. When Alzheimer's related genes are expressed in a tissue- and age-dependent manner using the bipartite GAL4-UAS system, flies exhibit a number of pathological hallmarks and behavioral impairments found in human patients with Alzheimer's disease including age dependent neurodegeneration, the accumulation of β-amyloid plaques, synaptic deficits, reduced life-span, and memory deficits (Brand and Perrimon, 1993; Greeve et al., 2004; Chakraborty et al., 2011; Mhatre et al., 2013, 2014; Blake et al., 2015). Surprisingly, while sleep disturbances are a common feature of Alzheimer's disease, very few studies have evaluated the impact of Alzheimer's related abnormal proteins on sleep in flies (Dissel et al., 2015a; Tabuchi et al., 2015; Gerstner et al., 2016).

    View all citing articles on Scopus
    1

    Present address: Baxter Lab for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.

    2

    Present address: Department of Animal Physiology, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland.

    View full text