. While NanoString data showed a similar expression trend for most of the transposable elements that were identified as differentially expressed in tau transgenic Drosophila by RNA-seq, some of the elements were not confirmed as differentially expressed by NanoString. These data reveal the limitations of each assay when analyzing transposable element transcripts and stress the importance of rigorous secondary validation. Since many members of the copia family are increased at the transcript level in both RNA-seq and NanoString analyses, it is speculated that gypsy-TRAP reporter activation is a result of copia insertion into the <i>ovo locus, rather than gypsy. Attempts to sequence de novo copia insertions within the <i>ovo locus in homogenates prepared from tau transgenic Drosophila heads resulted in a high frequency of mismatches, which is likely a result of the stochastic nature of transposable element insertion (Sun, 2018).
According to current understanding, cells have two layers of defense against potentially deleterious transposable element activation: transposable element transcription is limited by heterochromatin-mediated silencing, and transposable element transcripts are cleared from the cell by piRNA-mediated degradation. Both mechanisms of transposable element suppression were compromised in tauopathy. It is speculated that tau-induced heterochromatin decondensation facilitates active transcription of transposable elements and that tau-induced piwi and piRNA reduction allows those transcripts to persist. While these results are consistent with the effects of heterochromatin decondensation and piwi reduction on transposable element expression that have been reported in the Drosophila germline, these studies reveal a previously undocumented role for heterochromatin- and piRNA-mediated transposable element silencing in the brain. On the basis of studies in the germline reporting a direct interaction between piwi and HP140 and a requirement for Rhino, a member of the HP1 subfamily, for piRNA production, it is possible that a direct interaction between piwi and HP1 is also required to silence transposable elements in the brain (Sun, 2018).
Among upregulated transposable elements in human tauopathy, the human endogenous retrovirus (HERV) family, including HERV-K, was significantly over-represented. Elevated HERV-K transcripts are associated with amyotrophic lateral sclerosis (ALS)8 and many human cancers, including melanoma, breast cancer, germ cell tumors, renal cancer and ovarian cancer. A causal association between HERV-K and neuronal dysfunction has previously been established, as ectopic expression of HERV-K or the retroviral envelope protein that it encodes decreases synaptic activity and induces progressive motor dysfunction in mice. Antiretroviral reverse transcriptase inhibitors inhibit HERV-K activation in cultured cells and are now in clinical trials for the treatment of ALS. On the basis of the data presented in this study, reverse transcriptase inhibitors have significant potential as a therapeutic strategy for the treatment of neurodegenerative tauopathies, including Alzheimer's disease (Sun, 2018).
The ability of flamenco loss-of-function mutations to enhance tau-induced neurotoxicity and the ability of piwi overexpression, dietary restriction and inhibition of reverse transcriptase to reduce transposable element dysregulation and suppress tau-induced neurotoxicity suggest that tau-induced transposable element dysregulation is deleterious to neuronal survival. In addition to the detrimental effects of transposable element jumping, double-stranded RNAs produced by transposable element transcripts, including HERVs, can trigger a type I interferon response through the innate immune system. In light of the HERV increase in human tauopathy and the involvement of the innate immune response as a disease-promoting mechanism in Alzheimer's disease, it is tempting to speculate that expression of endogenous retroviruses in human tauopathy contributes to neuroinflammation, in addition to promoting genomic instability. In future studies, it will be important to investigate a potential effect of transposable element activation on the innate immune response in the context of tauopathy (Sun, 2018).
Younan, N. D., Chen, K. F., Rose, R. S., Crowther, D. C. and Viles, J. H. (2018). Prion protein stabilizes amyloid-beta (Abeta) oligomers and enhances Abeta neurotoxicity in a Drosophila model of Alzheimer disease. J Biol Chem. PubMed ID: 29887525
Abstract
The cellular prion protein (PrPC) can act as a cell-surface receptor for amyloid-beta (Abeta) peptide; however, a role for PrPC in the pathogenesis of Alzheimer's disease (AD) is contested. This study has expressed a range of Abeta isoforms and PrPC in the Drosophila brain. Co-expression of Abeta and PrPC significantly reduces the lifespan, disrupts circadian rhythms, and increases Abeta deposition in the fly brain. In contrast, under the same conditions, expression of Abeta or PrPC individually did not lead to these phenotypic changes. In vitro studies revealed that substoichiometric amounts of PrPC trap Abeta as oligomeric assemblies and fragment-preformed Abeta fibers. The ability of membrane-anchored PrPC to trap Abeta as cytotoxic oligomers at the membrane surface and fragment inert Abeta fibers, suggests a mechanism by which PrPC exacerbates Abeta deposition and pathogenic phenotypes in the fly, supporting a role for PrPC in AD. This study provides a second animal model linking PrPC expression with Abeta toxicity and supports a role for PrPC in AD pathogenesis. Blocking the interaction of Abeta and PrPC represents a potential therapeutic strategy (Younan, 2018).
Martin-Pena, A., Rincon-Limas, D. E. and Fernandez-Funez, P. (2018). Engineered Hsp70 chaperones prevent Abeta42-induced memory impairments in a Drosophila model of Alzheimer's disease. Sci Rep 8(1): 9915. PubMed ID: 29967544
Abstract
Proteinopathies constitute a group of diseases in which certain proteins are abnormally folded leading to aggregation and eventual cell failure. Most neurodegenerative diseases belong to protein misfolding disorders and, among them, Alzheimer's disease (AD) is the most prevalent. AD is characterized by accumulation of the amyloid-beta42 (Abeta42) peptide in the extracellular space. Hence, this study genetically engineered a molecular chaperone that was selectively delivered to this cellular location. It has been reported that the heat shock protein 70 (Hsp70) binds Abeta42 (see Drosophila Appl) preventing self-aggregation. This study employed two isoforms of the Hsp70, cytosolic and extracellular, to evaluate their potential protective effect against the memory decline triggered by extracellular deposition of Abeta42. Both Hsp70 isoforms significantly improved memory performance of flies expressing Abeta42, irrespective of their age or the level of Abeta42 load. Using olfactory classical conditioning, a Drosophila model of AD was established based on Abeta42 neurotoxicity and memory decline was monitored through aging. The onset of the memory impairment observed was proportional to the cumulative level of Abeta42 in the Drosophila brain. These data support the use of this Drosophila model of AD to further investigate molecules with a protective activity against Abeta42-induced memory loss, contributing to the development of palliative therapies for AD (Martin-Pena, 2018).
Talmat-Amar, Y., Arribat, Y. and Parmentier, M. L. (2018). Vesicular Axonal Transport is Modified In Vivo by Tau Deletion or Overexpression in Drosophila. Int J Mol Sci 19(3). PubMed ID: 29509687
Abstract
Structural microtubule associated protein Tau is found in high amount in axons and is involved in several neurodegenerative diseases. Although many studies have highlighted the toxicity of an excess of Tau in neurons, the in vivo understanding of the endogenous role of Tau in axon morphology and physiology is poor. Indeed, knock-out mice display no strong cytoskeleton or axonal transport phenotype, probably because of some important functional redundancy with other microtubule-associated proteins (MAPs). This study took advantage of the model organism Drosophila, which genome contains only one homologue of the Tau/MAP2/MAP4 family to decipher (endogenous) Tau functions. Tau depletion was found to lead to a decrease in microtubule number and microtubule density within axons, while Tau excess leads to the opposite phenotypes. Analysis of vesicular transport in tau mutants showed altered mobility of vesicles, but no change in the total amount of putatively mobile vesicles, whereas both aspects were affected when Tau was overexpressed. In conclusion, this study shows that loss of Tau in tau mutants not only leads to a decrease in axonal microtubule density, but also impairs axonal vesicular transport, albeit to a lesser extent compared to the effects of an excess of Tau (Talmat-Amar, 2018).
Guo, C., Jeong, H. H., Hsieh, Y. C., Klein, H. U., Bennett, D. A., De Jager, P. L., Liu, Z. and Shulman, J. M. (2018). Tau activates transposable elements in Alzheimer's disease. Cell Rep 23(10): 2874-2880. PubMed ID: 29874575
Abstract
Aging and neurodegenerative disease are characterized by genomic instability in neurons, including aberrant activation and mobilization of transposable elements (TEs). Integrating studies of human postmortem brain tissue and Drosophila melanogaster models, TE activation was investigated in association with Tau pathology in Alzheimer's disease (AD). Leveraging RNA sequencing from 636 human brains, differential expression was discovered for several retrotransposons in association with neurofibrillary tangle burden and highlight evidence for global TE transcriptional activation among the long interspersed nuclear element 1 and endogenous retrovirus clades. In addition, Tau-associated, active chromatin signatures were detected at multiple HERV-Fc1 genomic loci. To determine whether Tau is sufficient to induce TE activation, retrotransposons were profiled in Drosophila expressing human wild-type or mutant Tau throughout the brain. Heterogeneous response profiles were discovered, including both age- and genotype-dependent activation of TE expression by Tau. The results implicate TE activation and associated genomic instability in Tau-mediated AD mechanisms (Guo, 2018).
Panikker, P., Xu, S. J., Zhang, H., Sarthi, J., Beaver, M., Sheth, A., Akhter, S. and Elefant, F. (2018). Restoring Tip60 HAT/HDAC2 balance in the neurodegenerative brain relieves epigenetic transcriptional repression and reinstates cognition. J Neurosci. PubMed ID: 29654189
Abstract
Cognitive decline is a debilitating hallmark during pre-clinical stages of AD yet causes remain unclear. As histone acetylation homeostasis is critical for mediating epigenetic gene control throughout neuronal development, it was postulated that its misregulation contributes to cognitive impairment preceding AD pathology. This study shows that disruption of Tip60 HAT/HDAC2 homeostasis occurs early in the brain of an AD associated amyloid precursor protein (APP) Drosophila model and triggers epigenetic repression of neuroplasticity genes well before Abeta plaques form in male and female larvae. Repressed genes display enhanced HDAC2 binding and reduced Tip60 and histone acetylation enrichment. Increasing Tip60 in the AD associated APP brain restores Tip60 HAT/HDAC2 balance by decreasing HDAC2 levels, reverses neuroepigenetic alterations to activate synaptic plasticity genes, and reinstates brain morphology and cognition. Such Drosophila neuroplasticity gene epigenetic signatures are conserved in male and female mouse hippocampus and their expression and Tip60 function is compromised in hippocampus from AD patients. It is advocated that Tip60 HAT/HDAC2 mediated epigenetic gene disruption is a critical initial step in AD that is reversed by restoring Tip60 in the brain (Panikker, 2018).
Sarkar, A., Gogia, N., Glenn, N., Singh, A., Jones, G., Powers, N., Srivastava, A., Kango-Singh, M. and Singh, A. (2018). A soy protein Lunasin can ameliorate amyloid-beta 42 mediated neurodegeneration in Drosophila eye. Sci Rep 8(1): 13545. PubMed ID: 30202077
Abstract
Alzheimer's disease (AD), a fatal progressive neurodegenerative disorder, also results from accumulation of amyloid-beta 42 (Abeta42) plaques. These Abeta42 plaques trigger oxidative stress, abnormal signaling, which results in neuronal death by unknown mechanism(s). This study misexpressed high levels of human Abeta42 in the differentiating retinal neurons of the Drosophila eye, which results in the Alzheimer's like neuropathology. Using s transgenic model, a soy-derived protein Lunasin (Lun) was tested for a possible role in rescuing neurodegeneration in retinal neurons. Lunasin is known to have anti-cancer effect and reduces stress and inflammation. Misexpression of Lunasin by transgenic approach can rescue Abeta42 mediated neurodegeneration by blocking cell death in retinal neurons, and results in restoration of axonal targeting from retina to brain. Misexpression of Lunasin downregulates the highly conserved cJun-N-terminal Kinase (JNK) signaling pathway. Activation of JNK signaling can prevent neuroprotective role of Lunasin in Abeta42 mediated neurodegeneration. This neuroprotective function of Lunasin is not dependent on retinal determination gene cascade in the Drosophila eye, and is independent of Wingless and Decapentaplegic signaling pathways. Furthermore, Lunasin can significantly reduce mortality rate caused by misexpression of human Abeta42 in flies. These studies identified the novel neuroprotective role of Lunasin peptide, a potential therapeutic agent that can ameliorate Abeta42 mediated neurodegeneration by downregulating JNK signaling (Sarkar, 2018).
Jonson, M., Nystrom, S., Sandberg, A., Carlback, M., Michno, W., Hanrieder, J., Starkenberg, A., Nilsson, K. P. R., Thor, S. and Hammarstrom, P. (2018). Aggregated Abeta1-42 is selectively toxic for neurons, whereas glial cells produce mature fibrils with low toxicity in Drosophila. Cell Chem Biol 25(5):595-610. PubMed ID: 29657084
Abstract
The basis for selective vulnerability of certain cell types for misfolded proteins (MPs) in neurodegenerative diseases is largely unknown. This knowledge is crucial for understanding disease progression in relation to MPs spreading in the CNS. This issue was addressed in Drosophila by cell-specific expression of human Abeta1-42 associated with Alzheimer's disease. Expression of Abeta1-42 in various neurons resulted in concentration-dependent severe neurodegenerative phenotypes, and intraneuronal ring-tangle-like aggregates with immature fibril properties when analyzed by aggregate-specific ligands. Unexpectedly, expression of Abeta1-42 from a pan-glial driver produced a mild phenotype despite massive brain load of Abeta1-42 aggregates, even higher than in the strongest neuronal driver. Glial cells formed more mature fibrous aggregates, morphologically distinct from aggregates found in neurons, and was mainly extracellular. Theses findings implicate that Abeta1-42 cytotoxicity is both cell and aggregate morphotype dependent (Jonson, 2018).
Martin-Pena, A., Rincon-Limas, D. E. and Fernandez-Funez, P. (2017). Anti-Abeta single-chain variable fragment antibodies restore memory acquisition in a Drosophila model of Alzheimer's disease. Sci Rep 7(1): 11268. PubMed ID: 28900185
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder triggered by the accumulation of soluble assemblies of the amyloid-beta42 (Abeta42) peptide. Despite remarkable advances in understanding the pathogenesis of AD, the development of palliative therapies is still lacking. Engineered anti-Abeta42 antibodies are a promising strategy to stall the progression of the disease. Single-chain variable fragment (scFv) antibodies increase brain penetration and offer flexible options for delivery while maintaining the epitope targeting of full antibodies. This study examined the ability of two anti-Abeta scFv antibodies targeting the N-terminal (scFv9) and C-terminal (scFv42.2) regions of Abeta42 to suppress the progressive memory decline induced by extracellular deposition of Abeta42 in Drosophila. Using olfactory classical conditioning, both scFv antibodies were observed to significantly improve memory performance in flies expressing Abeta42 in the mushroom body neurons, which are intimately involved in the coding and storage of olfactory memories. The scFvs effectively restore memory at all ages, from one-day post-eclosion to thirty-day-old flies, proving their ability to prevent the toxicity of different pathogenic assemblies. These data support the application of this paradigm of Abeta42-induced memory loss in Drosophila to investigate the protective activity of Abeta42-binding agents in an AD-relevant functional assay (Martin-Pena, 2017).
Galasso, A., Cameron, C. S., Frenguelli, B. G. and Moffat, K. G. (2017). An AMPK-dependent regulatory pathway in tau-mediated toxicity. Biol Open [Epub ahead of print]. PubMed ID: 28808138
Abstract
Neurodegenerative tauopathies are characterized by accumulation of hyperphosphorylated tau aggregates primarily degraded by autophagy. The 5'AMP-activated protein kinase (AMPK) is expressed in most cells, including neurons. Alongside its metabolic functions, it is also known to be activated in Alzheimer's brains, phosphorylate tau, and be a critical autophagy activator. While stress conditions can result in AMPK activation enhancing tau-mediated toxicity, AMPK activation is not always concomitant with autophagic induction. This study analysed in Drosophila the impact of AMPK and autophagy on tau-mediated toxicity, recapitulating the AMPK-mediated tauopathy condition: increased tau phosphorylation, without corresponding autophagy activation. It was demonstrated that AMPK, binding to and phosphorylating tau at Ser-262, a site reported to facilitate soluble tau accumulation, affects its degradation. This phosphorylation results in exacerbation of tau toxicity and is ameliorated via rapamycin-induced autophagy stimulation. These findings support the development of combinatorial therapies effective at reducing tau toxicity targeting tau phosphorylation and AMPK-independent autophagic induction. The proposed in vivo tool represents an ideal readout to perform preliminary screening for drugs promoting this process (Galasso, 2017).
Zhang, X., Wang, W.A., Jiang, L.X., Liu, H.Y., Zhang, B.Z., Lim, N., Li, Q.Y. and Huang, F.D.(2017). Down-regulation of RBO-PI4KIIIα facilitates Aβ42 secretion and ameliorates neural deficits in Aβ42-expressing Drosophila. J Neurosci
[Epub ahead of print]. PubMed ID: 28424219
Abstract
Phosphoinositides and their metabolizing enzymes are involved in Aβ42
metabolism and Alzheimer's disease (AD) pathogenesis. In yeast and mammals, Eighty-five
requiring 3 (EFR3), whose Drosophila homolog is Rolling Blackout (RBO), forms a plasma membrane-localized protein complex
with phosphatidylinositol-4-kinase type IIIα (PI4KIIIα) and a scaffold protein to tightly control the
level of plasmalemmal phosphatidylinositol-4-phosphate (PI4P). This study
reports that RBO binds to Drosophila PI4KIIIα, and that in an
Aβ42-expressing Drosophila model, separate genetic reduction of
PI4KIIIα and RBO, or pharmacological inhibition of PI4KIIIα ameliorates
synaptic transmission deficit, climbing ability decline, and premature
death, and reduces neuronal accumulation of Aβ42. Moreover, RBO-PI4KIIIa
downregulation increases neuronal Aβ42 release, and PI4P facilitates
the assembly or oligomerization of Aβ42 in/on liposomes. These results
indicate that RBO-PI4KIIIa downregulation facilitates neuronal Aβ42
release and consequently reduces neuronal Aβ42 accumulation likely via
decreasing Aβ42 assembly in/on plasma membrane. This study suggests the
RBO-PI4KIIIα complex as a potential therapeutic target and PI4KIIIα
inhibitors as drug candidates for AD treatment (Zhang, 2017).
Lopez-Arias, B., Turiegano, E., Monedero, I., Canal, I. and Torroja, L. (2017). Presynaptic Abeta40 prevents synapse addition in the adult Drosophila neuromuscular junction. PLoS One 12(5): e0177541. PubMed ID: 28520784
Abstract
Complexity in the processing of the Amyloid Precursor Protein, which generates a mixture of βamyloid peptides, lies beneath the difficulty in understanding the etiology of Alzheimer's disease. Moreover, whether Aβ peptides have any physiological role in neurons is an unresolved question. By expressing single, defined Aβ peptides in Drosophila, specific effects can be discriminated in vivo. This study shows that in the adult neuromuscular junction (NMJ), presynaptic expression of Aβ40 hinders the synaptic addition that normally occurs in adults, yielding NMJs with an invariable number of active zones at all ages tested. A similar trend is observed for Aβ42 at young ages, but net synaptic loss occurs at older ages in NMJs expressing this amyloid species. In contrast, Aβ42arc produces net synaptic loss at all ages tested, although age-dependent synaptic variations are maintained. Inhibition of the PI3K synaptogenic pathway may mediate some of these effects, because western analyses show that Aβ peptides block activation of this pathway, and Aβ species-specific synaptotoxic effects persists in NMJs overgrown by over-expression of PI3K. Finally, individual Aβ effects are also observed when toxicity is examined by quantifying neurodegeneration and survival. These results suggest a physiological effect of Aβ40 in synaptic plasticity, and imply different toxic mechanisms for each peptide species (Lopez-Arias, 2017).
Frenkel-Pinter, M., Stempler, S., Tal-Mazaki, S., Losev, Y., Singh-Anand, A., Escobar-Alvarez, D., Lezmy, J., Gazit, E., Ruppin, E. and Segal, D. S.Altered protein glycosylation predicts Alzheimer's disease and modulates its pathology in disease model Drosophila. Neurobiol Aging. PubMed ID: 28552182
Abstract
The pathological hallmarks of Alzheimer's disease (AD) are pathogenic oligomers and fibrils of misfolded amyloidogenic proteins (e.g., beta-amyloid and hyper-phosphorylated tau in AD), which cause progressive loss of neurons in the brain and nervous system. In an analysis of available expression data sets this study indicates that many glycosylation-related genes are differentially expressed in brains of AD patients compared with healthy controls. The robust differences found enabled prediction of the occurrence of AD with remarkable accuracy in a test cohort and identification of a set of key genes whose expression determines this classification. Then the effect of reducing expression of homologs of 6 of these genes in was studied in transgenic Drosophila overexpressing human tau, a well-established invertebrate AD model. These experiments have led to the identification of glycosylation genes that may augment or ameliorate tauopathy phenotypes. These results indicate that OstDelta, l(2)not and beta4GalT7 are tauopathy suppressors, whereas pgnat5 and CG33303 are enhancers, of tauopathy. These results suggest that specific alterations in protein glycosylation may play a causal role in AD etiology (Frenkel-Pinter, 2017).
Feng, G., Pang, J., Yi, X., Song, Q., Zhang, J., Li, C., He, G. and Ping, Y. (2017). Down-regulation of KV4 channel in Drosophila mushroom body neurons contributes to Abeta42-induced courtship memory deficits. Neuroscience [Epub ahead of print]. PubMed ID: 28627422
Abstract
Accumulation of amyloid-β (Aβ) is widely believed to be an early event in the pathogenesis of Alzheimer's disease (AD). Kv4 is an A-type K+ channel, and previous work has shown that degradation of Kv4, induced by the β42 accumulation, may be a critical contributor to the hyperexcitability of neurons in a Drosophila AD model. This study used well-established courtship memory assay to investigate the contribution of the Kv4 channel to short-term memory (STM) deficits in the Aβ42-expressing AD model. Aβ42 over-expression in Drosophila leads to age-dependent courtship STM loss, which can be also induced by driving acute Aβ42 expression post-developmentally. Interestingly, mutants with eliminated Kv4-mediated A-type K+ currents (IA) by transgenically expressing dominant-negative subunit (DNKv4) phenocopied Aβ42 flies in defective courtship STM. Kv4 channels in mushroom body (MB) and projection neurons (PNs) were found to be required for courtship STM. Furthermore, the STM phenotypes can be rescued, at least partially, by restoration of Kv4 expression in Aβ42 flies, indicating the STM deficits could be partially caused by Kv4 degradation. In addition, IA is significantly decreased in MB neurons (MBNs) but not in PNs, suggesting Kv4 degradation in MBNs, in particular, plays a critical role in courtship STM loss in Aβ42 flies. These data highlight causal relationship between region-specific Kv4 degradation and age-dependent learning decline in the AD model, and provide a mechanism for the disturbed cognitive function in AD (Feng, 2017).
Wu, S. C., Cao, Z. S., Chang, K. M. and Juang, J. L. (2017). Intestinal microbial dysbiosis aggravates the progression of Alzheimer's disease in Drosophila. Nat Commun 8(1): 24. PubMed ID: 28634323
Abstract
Neuroinflammation caused by local deposits of Abeta42 in the brain is key for the pathogenesis and progression of Alzheimer's disease. However, inflammation in the brain is not always a response to local primary insults. Gut microbiota dysbiosis, which is recently emerging as a risk factor for psychiatric disorders, can also initiate a brain inflammatory response. It still remains unclear however, whether enteric dysbiosis also contributes to Alzheimer's disease. This study shows that in a Drosophila Alzheimer's disease model, enterobacteria infection exacerbated progression of Alzheimer's disease by promoting immune hemocyte recruitment to the brain, thereby provoking TNF-JNK mediated neurodegeneration. Genetic depletion of hemocytes attenuates neuroinflammation and alleviated neurodegeneration. It was further found that enteric infection increases the motility of the hemocytes, making them more readily attracted to the brain with an elevated oxidative stress status. This work highlights the importance of gut-brain crosstalk as a fundamental regulatory system in modulating Alzheimer's disease neurodegeneration. Emerging evidence suggests that gut microbiota influences immune function in the brain and may play a role in neurological diseases. This study offers in vivo evidence from a Drosophila model that supports a role for gut microbiota in modulating the progression of Alzheimer's disease (Wu, 2017).
Singh, S. K., Srivastav, S., Yadav, A. K. and Srikrishna, S. (2017). Knockdown of APPL mimics transgenic Abeta induced neurodegenerative phenotypes in Drosophila. Neurosci Lett 648: 8-13. PubMed ID: 28336338
Abstract
A variety of Drosophila mutant lines have been established as potential disease-models to study various disease mechanisms including human neurodegenerative diseases like Alzheimer's disease (AD), Huntington's disease (HD) and Parkinson's disease (PD). The evolutionary conservation of APP (Amyloid Precursor Protein) and APPL (Amyloid Precursor Protein-Like) and the comparable detrimental effects caused by their metabolic products strongly implies the conservation of their normal physiological functions. In view of this milieu, a comparative analysis on the pattern of neurodegenerative phenotypes between Drosophila APPL-RNAi line and transgenic Drosophila line expressing eye tissue specific human Aβ (Amyloid β) was undertaken. The results clearly show that Drosophila APPL-RNAi largely mimics transgenic Abeta in various phenotypes which include eye degeneration, reduced longevity and motor neuron deficit functions, etc. The ultra-structural morphological pattern of eye degeneration was confirmed by scanning electron microscopy. Further, a comparative study on longevity and motor behaviour between Abeta expressing and APPL knockdown lines revealed similar kind of behavioural deficit and longevity phenotypes. Therefore, it is suggested that APPL-knockdown can be used as an alternative approach to study neurodegenerative diseases in the fly model. This is the first report showing comparable phenotypes between APPL and Abeta in AD model of Drosophila (Singh, 2017).
Bergkvist, L., Sandin, L., Kagedal, K. and Brorsson, A. C. (2016). AβPP processing results in greater toxicity per amount of Aβ1-42 than individually expressed and secreted Aβ1-42 in Drosophila melanogaster. Biol Open [Epub ahead of print]. PubMed ID: 27387531
Abstract
The aggregation of the amyloid-β (Aβ; see Drosophila Appl) peptide into fibrillar deposits has long been considered the key neuropathological hallmark of Alzheimer's disease (AD). Aβ peptides are generated from proteolytic processing of the transmembrane Aβ precursor protein (AβPP) via sequential proteolysis through the β-secretase activity of β-site AβPP-cleaving enzyme (BACE1) and by the intramembranous enzyme γ-secretase. For over a decade, Drosophila melanogaster has been used as a model organism to study AD, and two different approaches have been developed to investigate the toxicity caused by AD-associated gene products in vivo. In one model, the Aβ peptide is directly over-expressed fused to a signal peptide, allowing secretion of the peptide into the extracellular space. In the other model, human AβPP is co-expressed with human BACE1, resulting in production of the Aβ peptide through the processing of AβPP by BACE1 and by endogenous fly γ-secretase. This study consisted of a parallel study of flies that expressed the Aβ1-42 peptide alone or that co-expressed AβPP and beta-secretase 1 (BACE1). Toxic effects (assessed by eye phenotype, longevity and locomotor assays) and levels of the Aβ1-42, Aβ1-40 and Aβ1-38 peptides were examined. The data reveal that the toxic effect per amount of detected Aβ1-42 peptide was higher in the flies co-expressing AβPP and BACE1 than in the Aβ1-42-expressing flies, and that the co-existence of Aβ1-42 and Aβ1-40 in the flies co-expressing AβPP and BACE1 could be of significant importance to the neurotoxic effect detected in these flies. Thus, the toxicity detected in these two fly models seems to have different modes of action and is highly dependent on how and where the peptide is generated rather than on the actual level of the Aβ1-42 peptide in the flies. This is important knowledge that needs to be taken into consideration when using Drosophila models to investigate disease mechanisms or therapeutic strategies in AD research (Bergkvist, 2016).
Go to top
Arnes, M., Casas-Tinto, S., Malmendal, A. and Ferrus, A. (2017)(2017). Amyloid beta42 peptide is toxic to non-neural cells in Drosophila yielding a characteristic metabolite profile and the effect can be suppressed by PI3K. Biol Open 6(11): 1664-1671. PubMed ID: 29141953
Abstract
The human Abeta42 peptide is associated with Alzheimer's disease through its deleterious effects in neurons. Expressing the human peptide in adult Drosophila in a tissue- and time-controlled manner, this study shows that Abeta42 is also toxic in non-neural cells, neurosecretory and epithelial cell types in particular. This form of toxicity includes the aberrant signaling by Wingless morphogen leading to the eventual activation of Caspase 3. Preventing Caspase 3 activation by means of p53 keeps epithelial cells from elimination but maintains the Abeta42 toxicity yielding more severe deleterious effects to the organism. Metabolic profiling by nuclear magnetic resonance (NMR) of adult flies at selected ages post Abeta42 expression onset reveals characteristic changes in metabolites as early markers of the pathological process. All morphological and most metabolic features of Abeta42 toxicity can be suppressed by the joint overexpression of PI3K (Arnes, 2017).
Go to top
Malmanche, N., Dourlen, P., Gistelinck, M., Demiautte, F., Link, N., Dupont, C., Vanden Broeck, L., Werkmeister, E., Amouyel, P., Bongiovanni, A., Bauderlique, H., Moechars, D., Royou, A., Bellen, H. J., Lafont, F., Callaerts, P., Lambert, J. C. and Dermaut, B. (2017). Developmental expression of 4-Repeat-Tau induces neuronal aneuploidy in Drosophila tauopathy models . Sci Rep 7: 40764. PubMed ID: 28112163
Abstract
Tau-mediated neurodegeneration in Alzheimer's disease and tauopathies is generally assumed to start in a normally developed brain. However, several lines of evidence suggest that impaired Tau isoform expression during development could affect mitosis and ploidy in post-mitotic differentiated tissue. Interestingly, the relative expression levels of Tau isoforms containing either 3 (3R-Tau) or 4 repeats (4R-Tau) play an important role both during brain development and neurodegeneration. This study used genetic and cellular tools to study the link between 3R and 4R-Tau isoform expression, mitotic progression in neuronal progenitors and post-mitotic neuronal survival. The results illustrated that the severity of Tau-induced adult phenotypes depends on 4R-Tau isoform expression during development. As recently described, a mitotic delay was observed in 4R-Tau expressing cells of larval eye discs and brains. Live imaging revealed that the spindle undergoes a cycle of collapse and recovery before proceeding to anaphase. Furthermore, a high level of aneuploidy was found in post-mitotic differentiated tissue. Finally, it was shown that overexpression of wild type and mutant 4R-Tau isoform in neuroblastoma SH-SY5Y cell lines is sufficient to induce monopolar spindles. Taken together, these results suggested that neurodegeneration could be in part linked to neuronal aneuploidy caused by 4R-Tau expression during brain development (Malmanche, 2017).
Go to top
Zhang, B., Li, Q., Chu, X., Sun, S. and Chen, S. (2016). Salidroside reduces tau hyperphosphorylation via up-regulating GSK-3β phosphorylation in a tau transgenic Drosophila model of Alzheimer's disease. Transl Neurodegener 5: 21. PubMed ID: 27933142
Abstract
Alzheimer's disease (AD) is an age-related and progressive neurodegenerative disease that causes substantial public health care burdens. Intensive efforts have been made to find effective and safe treatment against AD. The plant product Salidroside (Sal) is the main effective component of Rhodiola rosea L., which has several pharmacological activities. The objective of this study was to investigate the efficacy of Sal in the treatment of AD transgenic Drosophila and the associated mechanisms. Microtubule associated protein tau transgenic Drosophila line (TAU) was used in which tau protein is expressed in the central nervous system and eyes by the Gal4/UAS system. After feeding flies with Sal, the lifespan and locomotor activity were recorded. The appearance of vacuoles in the mushroom body was examined using immunohistochemistry, and the levels of total glycogen synthase kinase 3β (t-GSK-3β), phosphorylated GSK-3β (p-GSK-3β), t-tau and p-tau was detected in the brain by western blot analysis. The results showed that the longevity was improved in salidroside-fed Drosophila groups as well as the locomotor activity. Less vacuoles in the mushroom body, upregulated level of p-GSK-3β and downregulated p-tau were detected following Sal treatment. These data presented the evidence that Sal was capable of reducing the neurodegeneration in tau transgenic Drosophila and inhibiting neuronal loss. The neuroprotective effects of Sal were associated with its up-regulation of the p-GSK-3β and down-regulation of the p-tau (Zhang, 2016).
Go to top
Fernandez-Funez, P., Sanchez-Garcia, J., de Mena, L., Zhang, Y., Levites, Y., Khare, S., Golde, T. E. and Rincon-Limas, D. E. (2016). Holdase activity of secreted Hsp70 masks amyloid-β42 neurotoxicity in Drosophila. Proc Natl Acad Sci U S A 113: E5212-5221. PubMed ID: 27531960
Abstract
Alzheimer's disease (AD) is the most prevalent of a large group of related proteinopathies for which there is currently no cure. This study used Drosophila to explore a strategy to block Aβ42 neurotoxicity through engineering of the Heat shock protein 70 (Hsp70), a chaperone that has demonstrated neuroprotective activity against several intracellular amyloids. To target its protective activity against extracellular Aβ42, a signal peptide was added to Hsp70. This secreted form of Hsp70 (secHsp70) suppresses Aβ42 neurotoxicity in adult eyes, reduces cell death, protects the structural integrity of adult neurons, alleviates locomotor dysfunction, and extends lifespan. SecHsp70 binding to Aβ42 through its holdase domain is neuroprotective, but its ATPase activity is not required in the extracellular space. Thus, the holdase activity of secHsp70 masks Aβ42 neurotoxicity by promoting the accumulation of nontoxic aggregates. Combined with other approaches, this strategy may contribute to reduce the burden of AD and other extracellular proteinopathies (Fernandez-Funez, 2016).
Go to top
Niccoli, T., Cabecinha, M., Tillmann, A., Kerr, F., Wong, C.T., Cardenes, D., Vincent, A.J., Bettedi, L., Li, L., Grönke, S. Dols, J. and Partridge, L. (2016). Increased glucose transport into neurons rescues Aβ toxicity in Drosophila. Curr Biol [Epub ahead of print]. PubMed ID: 27524482
Abstract
Glucose hypometabolism is a prominent feature of the brains of patients with Alzheimer's disease (AD). Disease progression is associated with a reduction in glucose transporters in both neurons and endothelial cells of the blood-brain barrier. However, whether increasing glucose transport into either of these cell types offers therapeutic potential remains unknown. Using an adult-onset Drosophila model of Aβ (amyloid beta) toxicity, this study shows that genetic overexpression of a glucose transporter specifically in neurons, rescues lifespan, behavioral phenotypes, and neuronal morphology. This amelioration of Aβ toxicity is associated with a reduction in the protein levels of the unfolded protein response (UPR) negative master regulator Grp78 and an increase in the UPR. Genetic downregulation of Grp78 activity also protects against Aβ toxicity, confirming a causal effect of its alteration on AD-related pathology. Metformin, a drug that stimulates glucose uptake in cells, mimics these effects, with a concomitant reduction in Grp78 levels and rescue of the shortened lifespan and climbing defects of Aβ-expressing flies. These findings demonstrate a protective effect of increased neuronal uptake of glucose against Aβ toxicity and highlight Grp78 as a novel therapeutic target for the treatment of AD (Niccoli, 2016).
Go to top
Ray, A., Speese, S. D. and Logan, M. A. (2017) (2017). Glial Draper rescues Abeta toxicity in a Drosophila model of Alzheimer's Disease. J Neurosci 37(49):11881-11893. PubMed ID: 29109235
Abstract
Pathological hallmarks of Alzheimer's disease (AD) include amyloid-beta (Abeta) plaques, neurofibrillary tangles, and reactive gliosis. Glial cells offer protection against AD by engulfing extracellular Abeta peptides, but the repertoire of molecules required for glial recognition and destruction of Abeta are still unclear. This study shows that the highly conserved glial engulfment receptor Draper/MEGF10 provides neuroprotection in an AD model of Drosophila (both sexes). Neuronal expression of human Abeta42arc in adult flies results in robust Abeta accumulation, neurodegeneration, locomotor dysfunction, and reduced lifespan. Notably, all of these phenotypes are more severe in draper mutant animals, while enhanced expression of glial Draper reverses Abeta accumulation, as well as behavioral phenotypes. Stat92E, c-Jun N-terminal Kinase (JNK)/AP-1 signaling, and expression of matrix metalloproteinase-1 (Mmp1) are activated downstream of Draper in glia in response to Abeta42arc exposure. Furthermore, Abeta42-induced upregulation of the phagolysosomal markers Atg8 and p62 was notably reduced in draper mutant flies. Based on these findings, it is proposed that glia clear neurotoxic Abeta peptides in the AD model Drosophila brain through a Draper/STAT92E/JNK cascade that may be coupled to protein degradation pathways such as autophagy or more traditional phagolysosomal destruction methods (Ray, 2017).
Go to top
Vivien Chiu, W. Y., Koon, A. C., Ki Ngo, J. C., Edwin Chan, H. Y. and Lau, K. F. (2017) (2017). GULP1/CED-6 ameliorates amyloid-beta toxicity in a Drosophila model of Alzheimer's disease. Oncotarget 8(59): 99274-99283. PubMed ID: 29245900
Abstract
Amyloidogenic processing of APP (see Drosophila Appl) by beta- and gamma-secretases leads to the generation of amyloid-beta peptide (Abeta), and the accumulation of Abeta in senile plaques is a hallmark of Alzheimer's disease (AD). Understanding the mechanisms of APP processing is therefore paramount. Increasing evidence suggests that APP intracellular domain (AICD) interacting proteins influence APP processing. This study characterized the overexpression of AICD interactor GULP1 in a Drosophila AD model expressing human BACE and APP695. Transgenic GULP1 significantly lowered the levels of both Abeta1-40 and Abeta1-42 without decreasing the BACE and APP695 levels. Overexpression of GULP1 also reduced APP/BACE-mediated retinal degeneration, rescued motor dysfunction and extended longevity of the flies. These results indicate that GULP1 regulate APP processing and reduce neurotoxicity in a Drosophila AD model (Vivien Chiu, 2017).
Go to top
Oka, M., Fujisaki, N., Maruko-Otake, A., Ohtake, Y., Shimizu, S., Saito, T., Hisanaga, S. I., Iijima, K. M. and Ando, K. (2017). Ca2+/calmodulin-dependent protein kinase II promotes neurodegeneration caused by tau phosphorylated at Ser262/356 in a transgenic Drosophila model of tauopathy. J Biochem 162(5): 335-342. PubMed ID: 28992057
Abstract
Abnormal deposition of the microtubule-associated protein tau is a common pathological feature of multiple neurodegenerative diseases, including Alzheimer's disease (AD), and plays critical roles in their pathogenesis. Disruption of calcium homeostasis and the downstream kinase Ca2+/calmodulin-dependent protein kinase II (CaMKII) coincides with pathological phosphorylation of tau in AD brains. However, it remains unclear whether and how dysregulation of CaMKII affects tau toxicity. Using a Drosophila model, it was found that CaMKII promotes neurodegeneration caused by tau phosphorylated at the AD-associated sites Ser262/356. Overexpression of CaMKII promoted, while RNA-mediated knockdown of CaMKII and inhibition of CaMKII activity by expression of an inhibitory peptide suppressed, tau-mediated neurodegeneration. Blocking tau phosphorylation at Ser262/356 by alanine substitutions suppressed promotion of tau toxicity by CaMKII, suggesting that tau phosphorylation at these sites is required for this phenomenon. However, neither knockdown nor overexpression of CaMKII affected tau phosphorylation levels at Ser262/356, suggesting that CaMKII is not directly involved in tau phosphorylation at Ser262/356 in this model. These results suggest that a pathological cascade of events, including elevated levels of tau phosphorylated at Ser262/356 and aberrant activation of CaMKII, work in concert to promote tau-mediated neurodegeneration (OKA, 2017).
Go to top
Moore, B. D., Martin, J., de Mena, L., Sanchez, J., Cruz, P. E., Ceballos-Diaz, C., Ladd, T. B., Ran, Y., Levites, Y., Kukar, T. L., Kurian, J. J., McKenna, R., Koo, E. H., Borchelt, D. R., Janus, C., Rincon-Limas, D., Fernandez-Funez, P. and Golde, T. E. (2018). Short Abeta peptides attenuate Abeta42 toxicity in vivo. J Exp Med 215(1): 283-301. PubMed ID: 29208777
Abstract
Processing of amyloid-beta (Abeta) precursor protein (APP) by gamma-secretase produces multiple species of Abeta: Abeta40, short Abeta peptides (Abeta37-39), and longer Abeta peptides (Abeta42-43). gamma-Secretase modulators, a class of Alzheimer's disease therapeutics, reduce production of the pathogenic Abeta42 but increase the relative abundance of short Abeta peptides. To evaluate the pathological relevance of these peptides, this study expressed Abeta36-40 and Abeta42-43 in Drosophila melanogaster to evaluate inherent toxicity and potential modulatory effects on Abeta42 toxicity. In contrast to Abeta42, the short Abeta peptides were not toxic and, when coexpressed with Abeta42, were protective in a dose-dependent fashion. In parallel, the effects were explored of recombinant adeno-associated virus-mediated expression of Abeta38 and Abeta40 in mice. When expressed in nontransgenic mice at levels sufficient to drive Abeta42 deposition, Abeta38 and Abeta40 did not deposit or cause behavioral alterations. These studies indicate that treatments that lower Abeta42 by raising the levels of short Abeta peptides could attenuate the toxic effects of Abeta42 (Moore, 2018).
Go to top
Lee, B. I., Suh, Y. S., Chung, Y. J., Yu, K. and Park, C. B. (2017). Shedding light on Alzheimer's beta-Amyloidosis: Photosensitized methylene blue inhibits self-assembly of beta-amyloid peptides and disintegrates their aggregates. Sci Rep 7(1): 7523. PubMed ID: 28790398
Abstract
Abnormal aggregation of beta-amyloid (Abeta) peptides is a major hallmark of Alzheimer's disease (AD). In spite of numerous attempts to prevent the beta-amyloidosis, no effective drugs for treating AD have been developed to date. Among many candidate chemicals, methylene blue (MB) has proved its therapeutic potential for AD in a number of in vitro and in vivo studies; but the result of recent clinical trials performed with MB and its derivative was negative. In this study, with the aid of multiple photochemical analyses, it is reported that photoexcited MB molecules can block Abeta42 aggregation in vitro. Furthermore, an in vivo study using Drosophila AD model demonstrates that photoexcited MB is highly effective in suppressing synaptic toxicity, resulting in a reduced damage to the neuromuscular junction (NMJ), an enhanced locomotion, and decreased vacuole in the brain. The hindrance effect is attributed to Abeta42 oxidation by singlet oxygen (1O2) generated from photoexcited MB. Finally, it was shown that photoexcited MB possess a capability to disaggregate the pre-existing Abeta42 aggregates and reduce Abeta-induced cytotoxicity. This work suggests that light illumination can provide an opportunity to boost the efficacies of MB toward photodynamic therapy of AD in future (Lee, 2017).
Yang, C. N., Wu, M. F., Liu, C. C., Jung, W. H., Chang, Y. C., Lee, W. P., Shiao, Y. J., Wu, C. L., Liou, H. H., Lin, S. K. and Chan, C. C. (2017). Differential protective effects of connective tissue growth factor against Abeta neurotoxicity on neurons and glia. Hum Mol Genet 26(20): 3909-3921. PubMed ID: 29016849
Abstract
Impaired clearance of amyloid-beta peptide (Abeta; see Drosophila Appl) leads to abnormal extracellular accumulation of this neurotoxic protein that drives neurodegeneration in sporadic Alzheimer's disease (AD). Connective tissue growth factor (CTGF/CCN2) expression is elevated in plaque-surrounding astrocytes in AD patients. However, the role of CTGF in AD pathogenesis remains unclear. This study characterized the neuroprotective activity of CTGF. CTGF facilitates Abeta uptake and subsequent degradation within primary glia and neuroblastoma cells. CTGF enhanced extracellular Abeta degradation via membrane-bound matrix metalloproteinase-14 (MMP14) in glia and extracellular MMP13 in neurons. In the brain of a Drosophila AD model, glial-expression of CTGF reduced Abeta deposits, improved locomotor function, and rescued memory deficits. Neuroprotective potential of CTGF against Abeta42-induced photoreceptor degeneration was disrupted through silencing MMPs. Therefore, CTGF may represent a node for potential AD therapeutics as it intervenes in glia-neuron communication via specific MMPs to alleviate Abeta neurotoxicity in the central nervous system.
Go to top
Ando, K., Maruko-Otake, A., Ohtake, Y., Hayashishita, M., Sekiya, M. and Iijima, K. M. (2016). 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. PLoS Genet 12: e1005917. PubMed ID: 27023670
Abstract
To prevent the cascade of events leading to neurodegeneration in Alzheimer's disease (AD), it is essential to elucidate the mechanisms underlying the initial events of tau mismetabolism. In this study, using transgenic Drosophila co-expressing human tau and Aβ, tau phosphorylation at AD-related Ser262/356 stabilized microtubule-unbound tau was found 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 (Ando, 2016).
Go to top
Gerstner, J. R., Lenz, O., Vanderheyden, W. M., Chan, M. T., Pfeiffenberger, C. and Pack, A. I. (2016). Amyloid-β induces sleep fragmentation that is rescued by fatty acid binding proteins in Drosophila. J Neurosci Res [Epub ahead of print]. PubMed ID: 27320125
Abstract
Disruption of sleep/wake activity in Alzheimer's disease (AD) patients significantly affects their quality of life and that of their caretakers and is a major contributing factor for institutionalization. Levels of amyloid-β (Aβ; see Drosophila Appl) have been shown to be regulated by neuronal activity and to correlate with the sleep/wake cycle. Whether consolidated sleep can be disrupted by Aβ alone is not well understood. It was hypothesized that Aβ42 can increase wakefulness and disrupt consolidated sleep. This study shows that flies expressing the human Aβ42 transgene in neurons have significantly reduced consolidated sleep compared with control flies. Fatty acid binding proteins (Fabp) are small hydrophobic ligand carriers that have been clinically implicated in AD. Aβ42 flies that carry a transgene of either the Drosophila Fabp or the mammalian brain-type Fabp show a significant increase in nighttime sleep and long consolidated sleep bouts, rescuing the Aβ42-induced sleep disruption. These studies suggest that alterations in Fabp levels and/or activity may be associated with sleep disturbances in AD. Future work to determine the molecular mechanisms that contribute to Fabp-mediated rescue of Aβ42-induced sleep loss will be important for the development of therapeutics in the treatment of AD (Gerstner, 2016).
Go to top
Burnouf, S., Grönke, S., Augustin, H., Dols, J., Gorsky, M.K., Werner, J., Kerr, F., Alic, N., Martinez, P. and Partridge, L. (2016). Deletion of endogenous Tau proteins is not detrimental in Drosophila. Sci Rep 6: 23102. PubMed ID: 26976084
Abstract
Human Tau (hTau) is a highly soluble and natively unfolded protein that binds to microtubules within neurons. Its dysfunction and aggregation into insoluble paired helical filaments is involved in the pathogenesis of Alzheimer's disease (AD), constituting, together with accumulated β-amyloid (Aβ) peptides, a hallmark of the disease. Deciphering both the loss-of-function and toxic gain-of-function of hTau proteins is crucial to further understand the mechanisms leading to neurodegeneration in AD. As the fruit fly Drosophila melanogaster expresses Tau proteins (dTau) that are homologous to hTau, this study aimed to better comprehend dTau functions by generating a specific tau knock-out (KO) fly line using homologous recombination. It was observed that the specific removal of endogenous dTau proteins does not lead to overt, macroscopic phenotypes in flies. Indeed, survival, climbing ability and neuronal function are unchanged in tau KO flies. In addition, any overt positive or negative effect of dTau removal on human Aβ-induced toxicity were not found. Altogether, these results indicate that the absence of dTau proteins has no major functional impact on flies, and suggest that the tau KO strain is a relevant model to further investigate the role of dTau proteins in vivo, thereby giving additional insights into hTau functions (Burnouf, 2016).
Go to top
Farca Luna, A. J., Perier, M. and Seugnet, L. (2017). Amyloid precursor protein in Drosophila glia regulates sleep and genes Involved in glutamate recycling. J Neurosci 37(16): 4289-4300. PubMed ID: 28314820
Abstract
Amyloid precursor protein (App) plays a crucial role in Alzheimer's disease via the production and deposition of toxic β-amyloid peptides. App is heavily expressed in neurons, the focus of the vast majority of studies investigating its function. Meanwhile, almost nothing is known about App's function in glia, where it is also expressed, and can potentially participate in the regulation of neuronal physiology. This report investigated whether Appl, the Drosophila homolog of App, could influence sleep-wake regulation when its function is manipulated in glial cells. Appl inhibition in astrocyte-like and cortex glia resulted in higher sleep amounts and longer sleep bout duration during the night, while overexpression had the opposite effect. These sleep phenotypes were not the result of developmental defects, and were correlated with changes in expression in glutamine synthetase (GS) in astrocyte-like glia and in changes in the gap-junction component innexin2 in cortex glia. Downregulating both GS and innexin2, but not either one individually, resulted in higher sleep amounts, similarly to Appl inhibition. Consistent with these results, the expression of GS and innexin2 are increased following sleep deprivation, indicating that GS and innexin2 genes are dynamically linked to vigilance states. Interestingly, the reduction of GS expression and the sleep phenotype observed upon Appl inhibition could be rescued by increasing the expression of the glutamate transporter dEaat1. In contrast, reducing dEaat1 expression severely disrupted sleep. These results associate glutamate recycling, sleep, and a glial function for the App family proteins (Farca Luna, 2017).
Go to top
Gorsky, M. K., Burnouf, S., Dols, J., Mandelkow, E. and Partridge, L. (2016). Acetylation mimic of lysine 280 exacerbates human Tau neurotoxicity in vivo. Sci Rep 6: 22685. PubMed ID: 26940749
Abstract
Dysfunction and accumulation of the microtubule-associated human Tau (hTau) protein into intraneuronal aggregates is observed in many neurodegenerative disorders including Alzheimer's disease (AD). Reversible lysine acetylation has recently emerged as a post-translational modification that may play an important role in the modulation of hTau pathology. Acetylated hTau species have been observed within hTau aggregates in human AD brains and multi-acetylation of hTau in vitro regulates its propensity to aggregate. However, whether lysine acetylation at position 280 (K280) modulates hTau-induced toxicity in vivo is unknown. This study generated new Drosophila transgenic models of hTau pathology to evaluate the contribution of K280 acetylation to hTau toxicity, by analysing the respective toxicity of pseudo-acetylated (K280Q) and pseudo-de-acetylated (K280R) mutant forms of hTau. It was observed that mis-expression of pseudo-acetylated K280Q-hTau in the adult fly nervous system potently exacerbated fly locomotion defects and photoreceptor neurodegeneration. In addition, modulation of K280 influenced total hTau levels and phosphorylation without changing hTau solubility. Altogether, these results indicate that pseudo-acetylation of the single K280 residue is sufficient to exacerbate hTau neurotoxicity in vivo, suggesting that acetylated K280-hTau species contribute to the pathological events leading to neurodegeneration in AD (Gorsky, 2016).
Go to top
Sofola-Adesakin, O., Khericha, M., Snoeren, I., Tsuda, L. and Partridge, L. (2016). pGluAβ increases accumulation of Aβ in vivo and exacerbates its toxicity. Acta Neuropathol Commun 4: 109. PubMed ID: 27717375
Abstract
Several species of β-amyloid peptides (&Abeta;; see Drosophila Appl) exist as a result of differential cleavage from amyloid precursor protein (APP) to yield various C-terminal Aβ peptides. Several N-terminal modified Aβ peptides have also been identified in Alzheimer's disease (AD) brains, the most common of which is pyroglutamate-modified Aβ (AβpE3-42). AβpE3-42 peptide has an increased propensity to aggregate, appears to accumulate in the brain before the appearance of clinical symptoms of AD, and precedes Aβ1-42 deposition. Moreover, in vitro studies have shown that AβpE3-42 can act as a seed for full length Aβ1-42. This study characterized the Drosophila model of AβpE3-42 toxicity by expressing the peptide in specific sets of neurons using the GAL4-UAS system, and measuring different phenotypic outcomes. AβpE3-42 peptide was found to have an increased propensity to aggregate. Expression of AβpE3-42 in the neurons of adult flies led to behavioural dysfunction and shortened lifespan. Expression of AβpE3-42 constitutively in the eyes led to disorganised ommatidia, and activation of the c-Jun N-terminal kinase (JNK) signaling pathway. The eye disruption was almost completely rescued by co-expressing a candidate Aβ degrading enzyme, neprilysin2 (see Neprilysin 4). Furthermore, neprilysin2 was capable of degrading AβpE3-42. Also, the seeding hypothesis was tested for AβpE3-42 in vivo, and its effect on Aβ1-42 levels were measured. Aβ1-42 levels were significantly increased when Aβ1-42 and AβpE3-42 peptides were co-expressed. Furthermore, AβpE3-42 enhanced Aβ1-42 toxicity in vivo. These findings implicate AβpE3-42 as an important source of toxicity in AD, and suggest that its specific degradation could be therapeutic (Sofola-Adesakin, 2016).
Go to top
Xu, W., et al. (2016). Amyloid precursor protein-mediated endocytic pathway disruption induces axonal dysfunction and neurodegeneration. J Clin Invest [Epub ahead of print]. PubMed ID: 27064279
Abstract
The endosome/lysosome pathway is disrupted early in the course of both Alzheimer's disease (AD) and Down syndrome (DS); however, it is not clear how dysfunction in this pathway influences the development of these diseases. This study explored the cellular and molecular mechanisms by which endosomal dysfunction contributes to the pathogenesis of AD and DS. It was determined that full-length amyloid precursor protein (APP; see Drosophila Appl) and its β-C-terminal fragment (β-CTF) act though increased activation of Rab5 to cause enlargement of early endosomes and to disrupt retrograde axonal trafficking of nerve growth factor (NGF) signals. The functional impacts of APP and its various products were investigated in PC12 cells, cultured rat basal forebrain cholinergic neurons (BFCNs), and BFCNs from a mouse model of DS. The full-length wild-type APP (APPWT) and β-CTF both induced endosomal enlargement and disrupted NGF signaling and axonal trafficking. β-CTF alone induced atrophy of BFCNs that was rescued by the dominant-negative Rab5 mutant, Rab5S34N. Moreover, expression of a dominant-negative Rab5 construct markedly reduced APP-induced axonal blockage in Drosophila. Therefore, increased APP and/or β-CTF impact the endocytic pathway to disrupt NGF trafficking and signaling, resulting in trophic deficits in BFCNs. These data strongly support the emerging concept that dysregulation of Rab5 activity contributes importantly to early pathogenesis of AD and DS (Xu, 2016).
Go to top
Ping, Y., Hahm, E.T., Waro, G., Song,
Q., Vo-Ba, D.A., Licursi, A., Bao, H., Ganoe, L., Finch, K. and
Tsunoda, S. (2015). Linking Aβ42-induced
hyperexcitability to neurodegeneration, learning and motor
deficits, and a shorter lifespan in an Alzheimer's model. PLoS
Genet 11: e1005025. PubMed ID: 25774758
Abstract
Alzheimer’s disease (AD) is the most prevalent form of
dementia in the elderly. β-amyloid (Aβ) accumulation in
the brain is thought to be a primary event leading to eventual
cognitive and motor dysfunction in AD. Aβ has been shown to
promote neuronal hyperactivity, which is consistent with enhanced
seizure activity in mouse models and AD patients. Little, however,
is known about whether, and how, increased excitability
contributes to downstream pathologies of AD. This study shows that
overexpression of human Aβ42 in a Drosophila model
indeed induces increased neuronal activity. It was found that the
underlying mechanism involves the selective degradation of the
A-type K+ channel, Kv4.
An age-dependent loss of Kv4 leads to an increased probability of
AP firing. Interestingly, it was found that loss of Kv4 alone
results in learning and locomotion defects, as well as a shortened
lifespan.
To test whether the Aβ42-induced increase in neuronal
excitability contributes to, or exacerbates, downstream
pathologies, Kv4 was transgenically over-expressed to near
wild-type levels in Aβ42-expressing animals. It was shown
that restoration of Kv4 attenuates age-dependent learning and
locomotor deficits, slows the onset of neurodegeneration, and
partially rescues premature death seen in Aβ42-expressing
animals. The study concludes that Aβ42-induced hyperactivity
plays a critical role in the age-dependent cognitive and motor
decline of this Aβ42-Drosophila model, and possibly
in AD (Ping, 2015).
Highlights
- Aβ42 induces increased neuronal excitability in 9 days
old neuronal cultures.
- Kv4 current is selectively decreased by Aβ42 expression.
- Aβ42 induces degradation of Kv4 protein via a pathway
dependent on both the proteasome and lysosome.
- Transgenic restoration of Kv4 rescues hyperexcitability.
- Loss of Kv4 plays a critical role in Aβ42-induced
learning defects.
- Restoration of Kv4 levels rescues locomotor defects.
- Restoration of Kv4 slows Aβ42-induced neurodegeneration.
- Aβ42-induced loss of Kv4 contributes to premature death.
Discussion
Aβ-induced hyperexcitability is indeed intriguing, with
interesting implications especially for seizure-like activity and
epilepsy, which are potentially associated with AD. Little,
however, has been done previously to determine whether
Aβ-induced hyperactivity contributes to downstream behavioral
pathologies. Recent studies, however, suggest that neuronal
hyperactivity may precede neurological dysfunction and may be
improved by pharmacologically reducing activity. This study shows
that Kv4 channels are specifically down-regulated by Aβ42
expression, while other K+ currents (eg. Kv2 and Kv3) remain
unaltered in cultured neurons and in the intact brain. The
resulting increase in neuronal excitability is present in the
adult brain at an age (8 days AE) before the appearance of
locomotor (14–15 days AE) and learning defects (14 days AE),
and before the onset of neurodegeneration (25 days AE), supporting
the hypothesis that hyperactivity precedes and contributes to
these downstream pathologies. The study also shows that increasing
Kv4 channel levels in Aβ-expressing animals restores normal
excitability to neurons, and as a result, completely rescues
learning and locomotor defects, slows neurodegeneration, and
slightly increases lifespan. It is significant to note that the
expression of a UAS-GFP or UAS-CD8-GFP
transgene does not rescue any of these pathologies, suggesting
that any rescue effects by UAS-Kv4 are not simply due to
the introduction of another UAS target for GAL4 that
would dilute the expression of Aβ42; indeed, quantification
of Aβ42 is not any lower in Aβ42+Kv4 flies. In future
studies, it will be interesting to examine the temporal
requirement for reducing excitability with Kv4 expression; for
example, is early hyperexcitability more “toxic” to
the system than later stage hyperexcitability? (Ping, 2015).
Although specificity of rescue by Kv4 varies from one pathology
to another, the genetically engineered EKO channel that acts as a
general activity inhibitor does not ameliorate any of the
cognitive, motor, or survival deficits tested. This suggests that
general dampening of excitability is not sufficient to replace Kv4
loss. Kv1,
the other A-type K+ channel present in Drosophila,
however, is able to rescue Aβ42-induced locomotor
dysfunction, but, interestingly, not learning or premature death.
These results are consistent with the fact that Kv1 and Kv4 share
some, but certainly not all, biophysical properties. For example,
Kv4 channels have a much more hyperpolarized voltage-operating
range than Kv1 channels, making them much more likely to play
roles at subthreshold potentials. Also, while both Kv4 and Kv1
channels display fast inactivation, the inactivation rate is
voltage-independent for Kv4 channels and voltage-dependent for Kv1
channels. Finally, the subcellular localization of Kv4 and Kv1
channels are thought to be distinct, with Kv4 channels restricted
to dendrites and cell bodies, and Kv1 channels localized in axons
and nerve terminals (Ping, 2015).
The study also examined whether the loss of Kv4 function alone is
sufficient to lead to cognitive and motor pathologies. Previously,
it was shown that expression of a dominant-negative Kv4 subunit,
DNKv4, results in the elimination of the Kv4 current. Loss of Kv4
function leads to increased excitability and locomotor deficits.
In the present study, it was found that expression of DNKv4 also
induces learning defects and a shortened lifespan, consistent with
a key role for the Aβ42-induced reduction in Kv4 in these
downstream pathologies. In mammalian systems, Kv4.2 has been shown
to play a role in the induction of long-term potentiation (LTP),
and hippocampal dependent learning/memory defects. Loss of Kv4
function alone, however, does not induce any significant
neurodegeneration, suggesting that while Aβ42-induced loss of
Kv4 exacerbates degeneration, it is not sufficient to trigger
neurodegenerative pathway(s) (Ping, 2015).
Previous reports over the years have shown different effects of
Aβ on A-type K+ currents in vitro, with some identifying
decreases in A-type K+ currents (IA) and others reporting
increases in IA. Differences between these studies are likely to
be due to a variety of factors including the species of Aβ
tested (eg. Aβ1–40, Aβ1–42,
Aβ25–35; some studies finding clear differences with
different Aβ species, the cell type examined (eg. HEK cells,
hippocampal neurons, or cortical neurons), and the time course of
the effect (eg. from seconds to days in different studies). For
example, the Aβ species applied, the concentration used, and
time incubated with cells all affect the assembly state of
Aβ, which has also been proposed to have differential
downstream effects on K+ currents and excitability/activity. In
the future, it will be interesting to see how effects on Kv4
develop, and possibly change, throughout the assembly of Aβ42
from monomers to oligomers, protofibrils, and mature fibrils in
vivo (Ping, 2015).
Much remains to be understood about the mechanism by which Kv4
channels are lost in response to Aβ42 expression. In this
study, pharmacological and genetic approaches suggest a
degradation pathway for Kv4 that depends on both the proteasome
and lysosome, similar to the EGF receptor. This scenario is likely
to be complicated since previous studies have shown that Aβ
directly inhibits the proteasome, and that clearance of Aβ
depends on the proteasome. Further study is needed to understand
how Kv4 channels are targeted for turnover by Aβ42, and what
other component(s) are involved (Ping, 2015).
Further study is also needed to unravel specific mechanisms by
which Kv4 channels function in downstream Aβ42 pathologies.
For example, how does the loss of Kv4 exacerbate
neurodegeneration? One possibility is that cell death is induced
by an “excitotoxic” pathway due to excess Ca2+ entry,
and ultimately, necrosis. Interestingly, previous studies have
shown that Aβ42 induces an increase in various K+ currents
that are linked to cell death in vitro, consistent with evidence
that efflux of K+ is required as an early step in apoptosis. While
it is not clear how to reconcile these findings with ours, it does
seem that proper K+ homeostasis is critical for neuronal survival.
The role of Kv4 in lifespan, however, is complex, given that
neurodegeneration, learning/memory formation, and locomotor
activity all contribute to survival. The partial to full rescue of
multiple Aβ42-induced pathologies by Kv4, however,
underscores the importance of the loss of Kv4 in vivo and suggests
that Kv4 is a critical target of Aβ42 in this model, and
perhaps in AD (Ping, 2015).
Go to top
Blake, M.R., Holbrook, S.D.,
Kotwica-Rolinska, J., Chow, E.S., Kretzschmar, D. and
Giebultowicz, J.M. (2015). Manipulations of amyloid
precursor protein cleavage disrupt the circadian clock in aging
Drosophila. Neurobiol Dis 77: 117-126. PubMed ID: 25766673
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. This
study 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. It was shown 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. The study's data suggest
that behavioral rhythm disruption is not a product of APPL-derived
Aβ production but rather may be caused by a mechanism common
to both α and β-cleavage pathways. Specifically, it was
shown that increased production of the endogenous Drosophila
Amyloid Intracellular Domain (dAICD) causes disruption of
circadian rest-activity rhythms, while flies overexpressing
endogenous APPL maintain stronger circadian rhythms during aging.
In summary, this study offers a novel entry point toward
understanding the mechanism of circadian rhythm disruption in
Alzheimer's disease (Blake, 2015).
Highlights
- Over-expression of dBACE in clock cells accelerates aging
phenotypes and disrupts rest-activity rhythms.
- Neuronal over-expression of dBACE disrupts behavioral
rest-activity rhythms.
- Over-expression of dBACE dampens the cycling of PER in central
pacemaker neurons.
- Rest-activity rhythms are disrupted by KUZ over-expression.
- Expression of AICD disrupts rest-activity rhythms.
- dAICD is capable of entering the nucleus, but is not toxic to
central pacemaker neurons.
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, it is important to understand
relationships between AD and circadian rhythms at the cellular and
molecular levels. To address this question, this study examined
how manipulations of the fly ortholog of APP and its cleaving
enzymes affect endogenous rest-activity rhythms and clock
mechanism in Drosophila. Over-expression of dBACE was
found to disrupt behavioral rest-activity rhythms, and this effect
is most severe in aged flies suggesting an age-dependent
mechanism. Furthermore, dBACE expression resulted in dampened
oscillation of the core clock protein PER in central pacemaker
neurons, which are master regulators of rest activity rhythms.
Significantly reduced PER levels are observed in the sLNv and lLNv
neurons of age 50d flies expressing dBACE in all clock cells
(including glia), all neurons, or only in PDF-positive sLNv and
lLNv neurons. These data suggest that manipulation of APP-cleavage
by dBACE over-expression directly affects the oscillation of PER
protein in central pacemaker neurons in a cell-autonomous manner.
Since a functional clock mechanism in sLNv is necessary and
sufficient to maintain free running activity rhythms, reduced
oscillations of PER in these neurons could be responsible for the
loss of activity rhythms in age 50d flies. Importantly, the
decline in PER levels occurrs only in flies with manipulated
dBACE, not in old control flies. This is in agreement with earlier
findings that aging does not dampen PER oscillations in pacemaker
neurons of wild type flies, while it reduces clock oscillations in
peripheral clocks (Blake, 2015).
While this study reports that the loss of behavioral rhythms
after manipulation of dBACE is associated with reduced expression
of clock genes in the central pacemaker, other recent work shows
that expression of human Aβ peptides leads to disruption of
rest activity rhythms without interfering with PER oscillations in
the central pacemaker. Even strongly neurotoxic Aβ peptides,
such as Aβ42 arctic, do not cause rhythm disruption when
expressed in central pacemaker neurons; rather, pan-neuronal
expression is required. The fact that even the most neurotoxic
Aβ peptides are not capable of dampening PER oscillation in
pacemaker neurons suggests that Aβ production does not affect
clock oscillations and that it is not Aβ production that
causes the phenotype observed in this study upon over-expression
of dBACE. This was confirmed by expression of KUZ,
whose activity does not increase dAβ production; however, it
also leads to disruption of rest-activity rhythms. Similar rhythm
disruption by dBACE and KUZ suggests that an excess cleavage
product of both pathways might be responsible for the disruption.
Like in the mammalian APP cleavage pathway, in Drosophila
cleavage of APPL by KUZ or dBACE results in a C-terminal fragment
(CTF) that is subsequently cleaved by the ϒ-secretase
resulting in the production of dAICD. Indeed, it was shown that
expression of dAICD results in an age-dependent decline in
rhythmic locomotor activity. As with dBACE and KUZ expression,
dAICD expression causes weakening or complete loss of behavioral
rhythms while age-matched control flies remain highly rhythmic. In
this context, it is worth noting that α-secretase activators
are considered for clinical trials to reduce Aβ production in
AD patients. However, according to results in this study, this
could lead to disruptions of circadian rhythms and sleep patterns
thus negatively impacting the lives of patients and their
caretakers (Blake, 2015).
This study's data suggest that increased dAICD may be the
proximal cause of decay in rest-activity rhythms. The role of AICD
in AD is increasingly evident but poorly understood. AICD is able
to enter the nucleus and has been implicated in transcriptional
regulation that may affect cell death, neurite outgrowth and
neuronal excitability. Interestingly, transgenic mice expressing
AICD have increased activity of GSK-3, which in flies affects the
circadian clock. Over-expression of GSK-3
in Drosophila leads to altered circadian behavior by
hyper-phosphorylation of
TIMELESS (TIM), a key circadian protein which forms dimers
with PER that enter the nucleus and regulate the clock mechanism.
Of further interest, increased GSK-3 activity has been implicated
in AD, and in Drosophila, increased GSK-3 activity
mediates the toxicity of Aβ peptides (Blake, 2015).
Cleavage of APPL likely results in a significant decline in
intact APPL, and this could be detrimental as APPL has
neuroprotective effects. It was also recently shown that loss of
full-length APPL induces cognitive deficits in memory. This study
reports that flies over-expressing full-length APPL in central
pacemaker neurons maintain stronger behavioral rest-activity
rhythms during aging than control flies; however this effect is
not observed when APPL is expressed pan-neuronally. This could be
caused by negative effects of APPL when expressed in other
unspecified neurons, or could be related to driver strength.
Overall, the study suggests that the loss of full-length APPL
might negatively affect circadian behavior by way of the central
pacemaker neurons (Blake, 2015).
Over-expression of dAICD induces a severe phenotype, disrupting
rest-activity rhythms as early as age 5d when expressed in central
pacemaker neurons and by age 35d with pan-neuronal expression.
Taken together these results suggest that while loss of
full-length APPL by over-expression of its secretases might
negatively impact circadian behavior, the cleavage product dAICD
induces the most severe behavioral rest-activity disruption.
Interestingly, the observed effect is not likely a product of
neurodegeneration as it was previously shown that dAICD has no
effect on neurodegeneration, and this study shows that the
pacemaker cells appear intact in pdf > dAICD flies.
In addition, it was shown that dAICD, like the vertebrate AICD,
can be found in the nucleus. Therefore, this study suggests that
dAICD may directly or indirectly affect the expression of clock
genes. This offers a novel entry point toward understanding the
mechanism of circadian rhythm disruption in Alzheimer's disease
(Blake, 2015).
Go to top
Long, D.M., Blake, M.R., Dutta, S.,
Holbrook, S.D., Kotwica-Rolinska, J., Kretzschmar, D. and
Giebultowicz, J.M. (2014). Relationships between the
circadian system and Alzheimer's disease-like symptoms in Drosophila.
PLoS One 9: e106068. PubMed ID: 25171136
Abstract
Circadian clocks coordinate physiological, neurological, and
behavioral functions into circa 24 hour rhythms, and the molecular
mechanisms underlying circadian clock oscillations are conserved
from Drosophila to humans. Clock oscillations and
clock-controlled rhythms are known to dampen during aging;
additionally, genetic or environmental clock disruption leads to
accelerated aging and increased susceptibility to age-related
pathologies. Neurodegenerative diseases, such as Alzheimer's
disease (AD), are associated with a decay of circadian rhythms,
but it is not clear whether circadian disruption accelerates
neuronal and motor decline associated with these diseases. To
address this question, this study utilized transgenic Drosophila
expressing various Amyloid-β (Aβ) peptides, which are
prone to form aggregates characteristic of AD pathology in humans.
The study compared development of AD-like symptoms in adult flies
expressing Aβ peptides in the wild type background and in
flies with clocks disrupted via a null mutation in the clock gene
period
(per01). No significant differences were observed in
longevity, climbing ability and brain neurodegeneration levels
between control and clock-deficient flies, suggesting that loss of
clock function does not exacerbate pathogenicity caused by
human-derived Aβ peptides in flies. However, AD-like
pathologies were found to affect the circadian system in aging
flies. It was found that rest/activity rhythms are impaired in an
age-dependent manner. Flies expressing the highly pathogenic
arctic Aβ peptide show a dramatic degradation of these
rhythms in tune with their reduced longevity and impaired climbing
ability. At the same time, the central pacemaker remains intact in
these flies providing evidence that expression of Aβ peptides
causes rhythm degradation downstream from the central clock
mechanism (Long, 2014).
Highlights
- Lifespan reduction caused by Aβ peptides is not
exacerbated by the by loss of the clock gene period.
- Flies expressing Aβ42arc show similar motor decline and
neurodegeneration in clock-positive and clock-disrupted
backgrounds.
- Daily locomotor activity rhythms are impaired in aging flies
expressing different Aβ peptides.
- Rhythms in PER cycling continue in lateral neurons of
elav>Aβ42arc flies.
Discussion
Associations between AD and impaired daily rhythms are well
documented in humans, yet the causes and consequences of
AD-related loss of circadian sleep/activity rhythms have not been
teased apart. One of the unanswered questions is whether
age-related decline of the circadian system contributes to AD
progression. This study tested directly whether total arrhythmia
caused by mutation in the core clock gene per would
exacerbate AD-like phenotypes observed in an AD fly model. It was
shown that premature death, progressive locomotor deficits, and
vacuolization in the brain occurs with similar timing and
intensity in flies with genetically disrupted clock mechanism as
in control flies. Consistent with previous reports, the severity
of symptoms is proportional to the pathogenicity of the expressed
human Aβ fragments. However, within each genotype, symptoms
in clock-deficient flies are similar to those in clock-competent
flies. While this study's data show that disruption of the clock
via removal of the core clock repressor PER does not exacerbate AD
symptoms, it cannot be ruled out that disabling the positive clock
arm could be more detrimental. A recent report showed that loss of
the positive element BMAL1 causes brain neurodegeneration in mice.
It was previously demonstrated that the loss of per
accelerates death, locomotor impairments, and brain vacuolization
in neurodegeneration-prone sniffer and swiss cheese fly mutants.
However, the underlying molecular mechanism that mediates this
effect is not known. The AD model used in this study is based on
the expression of human Aβ peptides, which have been reported
to accumulate into insoluble forms in aging flies. Because the
disruption of the circadian clock does not affect the
pathogenicity of these peptides, the study assumes that it has no
effect on Aβ aggregation or clearance. In sum, this study's
data show that the molecular and behavioral arrhythmia
characteristic for per-null flies is not detrimental in
this AD fly model (Long, 2014).
However, the study shows that associations between AD and altered
behavioral rhythms, observed in humans and AD model mice, also
extend to fly AD models. Pan-neuronal expression of Aβ42
causes age-dependent impairment of circadian rest/activity
rhythms, such that a reduced fraction of 50-days old elav>Aβ42
flies remain rhythmic in constant darkness compared to controls. A
more dramatic disruption of circadian rhythms is observed in elav>Aβ42arc.
In LD, 5-day old flies of this genotype show bimodal activity with
an attenuated morning activity peak, while no activity peaks are
detected in 15-day old flies, rather they are active around the
clock, including nighttime when control flies have prolonged rest.
In another related study, a loss of locomotor activity rhythms in
elav>Aβ42arc flies even at young age was shown,
similar to findings in this study. Together, these results
demonstrate that AD model flies have rest/activity rhythm
degradation reminiscent of the behavioral degradation observed in
humans with AD (Long, 2014).
Loss of rest/activity rhythms in elav>Aβ42arc
flies formed the basis of investigation of the functional status
of central pacemaker neurons, which are necessary and sufficient
for the activity rhythms, at least in young flies.
Immunocytochemistry of PDF-positive pacemaker neurons sLNv and
lLNv shows the correct number and arborization pattern in elav>Aβ42arc
flies. Moreover these neurons show nuclear peak and trough of the
core clock protein PER indistinguishable from wild type flies.
Similar observations have been published earlier, and it was
additionally shown that even expression of the more pathogenic
tandem Aβ42 construct leaves molecular oscillations in
pacemaker neurons intact. Together, these data show dissociation
between functioning molecular pacemaker and disrupted circadian
coordination of rest/activity rhythms. This suggests that
behavioral rhythm degradation observed in humans and mouse AD
models may occur despite the presence of a functional central
clock. Importantly, strong body temperature rhythms have been
reported in AD patients again suggesting that the central clock
may be intact in AD. This is reminiscent of the situation in very
old flies and mammals, which show degradation of rest/activity
rhythms while their central pacemaker neurons continue to show
molecular oscillations (Long, 2014).
While AD-related degradation of behavioral rhythms is not caused
by malfunction of the central clock, other contributing factors
remain to be investigated. Aβ related arrhythmicity might be
due to non-cell-autonomous toxicity as focused expression of toxic
peptides in clock containing cells does not affect behavioral
rhythmicity, but expression outside of the pacemaker neurons may
affect their synaptic connections. Additionally, downstream
neuronal or humoral output pathways leading from the central
pacemaker network to the motor centers could be adversely affected
by Aβ aggregates. For example, recent studies reporting a
direct measurement of neuronal activity in elav>Aβ42arc
flies reveal increased latency and decreased response stability of
the pathways leading from the giant fiber system in the brain into
motor neurons of the thoracic ganglia. It is possible that
neuronal deficits of this kind can disable output pathways from
the central clock leading to fragmented rather than consolidated
sleep. This may lead to a vicious cycle as sleep deprivation
increases amyloid peptides in mice and Aβ aggregation
disrupts the sleep/wake cycle. As flies provide a powerful toolkit
to study both AD and circadian rhythms, studies at the
intersection of chronobiology and AD should help to provide
insights into the mechanisms underlying AD-related pathologies
(Long, 2014).
Go to top
Wang, X., Ma, Y., Zhao, Y., Chen, Y.,
Hu, Y., Chen, C., Shao, Y. and Xue, L. (2015). APLP1
promotes dFoxO-dependent cell death in Drosophila.
Apoptosis 20: 778-786. PubMed ID: 25740230
Abstract
The amyloid
precursor like protein-1 (APLP1), an engineered human gene
into the Drosophila genome, belongs to the amyloid precursor
protein family that also includes the Drosophila amyloid precursor
protein (APP) and the amyloid
precursor like protein-2 (APLP2). Though the three proteins
share similar structures and undergo the same cleavage processing
by α-, β- and γ-secretases, APLP1 shows divergent
subcellular localization from that of APP and APLP2, and thus, may
perform distinct roles in vivo. While extensive studies have been
focused on APP, which is implicated in the pathogenesis of
Alzheimer’s disease, the functions of APLP1 remain largely
elusive. This study reports that the expression of APLP1 in Drosophila
induces cell death and produces developmental defects in wing and
thorax. This function of APLP1 depends on the transcription factor
dFoxO, as the depletion of dFoxO abrogates APLP1-induced cell
death and adult defects. Consistently, APLP1 up-regulates the
transcription of dFoxO target hid
and reaper-two
well known pro-apoptotic genes. Thus, the present study provides
the first in vivo evidence that APLP1 is able to induce cell
death, and that FoxO is a crucial downstream mediator of
APLP1’s activity (Wang, 2015).
Highlights
- APLP1 induces caspase-dependent cell death in Drosophila.
- APLP1 up-regulates dFoxO target gene expression.
Discussion
Amyloid pecursor like protein-1(APLP1) is a mammalian paralog of
amyloid precursor protein (APP). While APP has been extensively
studied for its involvement in the Alzheimer’s disease, few
studies have been directed to APLP1 and its in vivo functions
remain largely unknown. This study investigated the in vivo
functions of APLP1 using Drosophila as a model organism.
It was found that ectopic expression of APLP1 induces cell death
and developmental defects in the nervous and non-nervous system.
Genetic studies characterize the transcription factor dFoxO as a
critical downstream factor that mediates APLP1’s activity,
for the depletion of dFoxO significantly suppresses APLP1-induced
cell death in larval discs and associated phenotypes in adults.
Further, it was shown that APLP1 is able to up-regulate the
transcription of dFoxO target genes hid and reaper
(Wang, 2015).
APLP1 has been reported to function mainly in the nervous system,
as high expression level of APLP1 is detected in the developing
central and peripheral nervous systems, yet a weak expression
signal of APLP1 is also observed in organs like heart, lung, liver
and kidney in mouse embryos, implying a role of APLP1 in the
development of non-neuronal tissues. Consistent with this
explanation, RNAi mediated knockdown of APLP1 in WI-38 and MCF7
cells dramatically reduced the proliferation of these cells. This
study showed that expression of APLP1 could induce cell death and
developmental defects in both neuronal and non-neuronal systems in
Drosophila, and thus, provides further evidence for the
function of APLP1 in non-neuronal cells (Wang, 2015).
Earlier studies have also shown that loss of APLP1 diminishes
stress induced apoptosis in neuroblastoma cells, whereas ectopic
expression of APLP1 moderately enhances cell death upon stress
stimulation. However, expression of APLP1 alone is not sufficient
to induce neuroblastoma cell death , suggesting APLP1 induces cell
death in a context dependent manner. Data from this study
demonstrates that APLP1 by itself is sufficient to induce cell
death. APLP1 has been reported to be a direct transcriptional
target of the p53 tumor suppressor, which suggests a possible
involvement of APLP1 in p53-induced cell death. p53 is known to
interact with the transcriptional factor FoxO, and MDM2 is known
to act downstream of p53 to promote FoxO ubiquitination and
degradation. In the present study, it was shown that FoxO mediates
APLP1-induced cell death. The exact relationship between APLP1,
FoxO and p53 in cell death will require further investigation.
Overall, this study highlights a novel function of APLP1 in
promoting FoxO-mediated cell death in vivo, which will shed light
on the role of APLP1 in mammalian cells (Wang, 2015).
Go to top
Omata, Y., Lim, Y.M., Akao, Y. and
Tsuda, L. (2014). Age-induced reduction of
autophagy-related gene expression is associated with onset of
Alzheimer's disease. Am J Neurodegener Dis 3: 134-142. PubMed ID:
25628964
Abstract
Aging is a major risk factor for Alzheimer's disease (AD).
Aggregation of amyloid beta (Aβ) in cerebral cortex and
hippocampus is a hallmark of AD. Many factors have been identified
as causative elements for onset and progression of AD; for
instance, tau seems to mediate the neuronal toxicity of Aβ,
and downregulation of macroautophagy (autophagy) is thought to be
a causative element of AD pathology. Expression of
autophagy-related genes is reduced with age, which leads to
increases in oxidative stress and aberrant protein accumulation.
This study found that expression of the autophagy-related genes atg1,
atg8a,
and atg18
in Drosophila melanogaster is regulated with aging as
well as their own activities. In addition, the level of atg18
is maintained by dfoxo (foxo) and dsir2
(sir2) activities in concert with aging. These results
indicate that some autophagy-related gene expression is regulated
by foxo/sir2-mediated aging processes. It was further
found that reduced autophagy activity correlates with late-onset
neuronal dysfunction caused by neuronal induction of Aβ.
These data support the idea that age-related dysfunction of
autophagy is a causative element in onset and progression of AD
(Omata, 2014).
Highlights
- Expression of autophagy-related genes is reduced with aging.
- Expression of atg18 is reduced in dfoxo
and dsir2 mutants.
- Initiation factor 4E-binding
protein (4E-BP) negatively regulates autophagy-related
gene expression.
- Aβ transgenic flies produce Aβ42 and exhibit reduced
locomotor activity.
- Expression of autophagy-related genes affects development of
AD.
- Expression of autophagy-related genes correlates with neuronal
toxicity caused by Aβ42.
Discussion
This study shows that expression of autophagy-related genes is
regulated by age-related signaling. dsir2 (a Drosophila
SIRT1 homolog) and dfoxo are required to maintain atg18
expression during aging, suggesting that, among autophagy-related
genes, this gene specifically is regulated by foxo/sir2
activity. Interestingly, aging seems to affect expression of all
autophagy-related genes tested, suggesting that aging and foxo/sir2
may act at different levels to regulate autophagy-related gene
expression (Omata, 2014).
Previous studies show that sir2, foxo and 4E-BP
are involved in regulating the Drosophila lifespan. Data
from this study, however, indicate that 4E-BP
antagonizes expression of autophagy-related genes. 4E-BP
is believed to be controlled by TOR
signaling. Therefore, the negative effect of 4E-BP
on autophagy-related gene expression may be mediated through the
effect of TOR signaling pathway, which also seems to antagonize
autophagy-related gene expression (Omata, 2014).
Autophagy is highly correlated with lysosomal activity, and the
autophagy-lysosome pathway is thought to be involved in many
cellular processes. Earlier studies indicate that lysosomal
activity affects expression of autophagy-related genes. The
lysosome nutrient sensing (LYNUS) machinery is responsible for
sensing whether there are sufficient nutrients. Under a sufficient
nutrient status, the mammalian target of rapamycin complex 1
(mTORC1, a member of the LYNUS machinery) phosphorylates
transcription factor EB (TFEB) on the lysosomal surface and
inhibits its nuclear localization. In this way, TFEB is unable to
induce expression of lysosomal and autophagy-related genes under
nutrient sufficient conditions. These results suggest that the
level of autophagy-related genes might be regulated by the state
of lysosome formation and autophagy itself. Here, expression of
autophagy-related genes is affected by the activity of other
autophagy-related genes as well as their own activity, suggesting
that auto-feedback regulation is part of the mechanism used to
maintain expression of autophagy-related genes in Drosophila
(Omata, 2014).
It was observed that reducing the expression of autophagy-related
genes strongly enhances the neuronal toxicity caused by Aβ
expression. Furthermore, reducing atg1 expression using
the Df(atg1)/+ heterozygote shows a more severe
enhancement of Aβ-dependent neuronal toxicity than reducing atg18
expression using the Df(atg18)/+ heterozygote.
Interestingly, atg1 also demonstrates strong
auto-feedback regulation, as reducing expression of atg1
results in further defects in expression of atg genes.
Therefore, it is possible that a drastic reduction in expression
of many atg genes may contribute to the neuronal
toxicity of Aβ42, and that aging and autophagy may be
determinants of AD onset (Omata, 2014).
Go to top
Saitoh, Y., Fujikake, N., Okamoto, Y.,
Popiel, H.A., Hatanaka, Y., Ueyama, M., Suzuki, M., Gaumer, S.,
Murata, M., Wada, K. and Nagai, Y. (2015). p62 plays a
protective role in the autophagic degradation of polyglutamine
protein oligomers in polyglutamine disease model flies. J Biol
Chem 290: 1442-1453. PubMed ID: 25480790
Abstract
Oligomer formation and accumulation of pathogenic proteins are key
events in the pathomechanisms of many neurodegenerative diseases,
such as Alzheimer disease, ALS, and the polyglutamine (polyQ)
diseases. The autophagy-lysosome degradation system may have
therapeutic potential against these diseases because it can
degrade even large oligomers. Although p62/sequestosome 1 plays a
physiological role in selective autophagy of ubiquitinated
proteins, whether p62 recognizes and degrades pathogenic proteins
in neurodegenerative diseases has remained unclear. This study
used Drosophila models of neurodegenerative diseases to
elucidate the role of p62 in such pathogenic conditions in vivo.
It was found that p62 predominantly co-localizes with cytoplasmic
polyQ protein aggregates in the MJDtr-Q78 polyQ disease model
flies. Loss of p62 function results in significant exacerbation of
eye degeneration in these flies. Immunohistochemical analyses
revealed enhanced accumulation of cytoplasmic aggregates by p62
knockdown in the MJDtr-Q78 flies, similarly to knockdown of
autophagy-related genes (Atgs). Knockdown of both p62
and Atgs did not show any additive effects in the
MJDtr-Q78 flies, implying that p62 function is mediated by
autophagy. Biochemical analyses showed that loss of p62 function
delays the degradation of the MJDtr-Q78 protein, especially its
oligomeric species. It was also found that loss of p62 function
exacerbates eye degeneration in another polyQ disease fly model as
well as in ALS model flies. The study therefore concludes that p62
plays a protective role against polyQ-induced neurodegeneration,
by the autophagic degradation of polyQ protein oligomers in vivo,
indicating its therapeutic potential for the polyQ diseases and
possibly for other neurodegenerative diseases (Saitoh, 2015).
Highlights
- p62 co-localizes with cytoplasmic polyQ protein aggregates.
- Loss of p62 function causes exacerbation of eye degeneration
in polyQ disease model flies.
- Loss of p62 function results in an increase in cytoplasmic
polyQ protein aggregates.
- Loss of autophagic function causes the exacerbation of eye
degeneration accompanied by the enhanced accumulation of
cytoplasmic polyQ protein aggregates.
- Protective role of p62 against polyQ protein toxicity is
dependent on autophagy.
- Loss of p62 function delays the degradation of the polyQ
protein in vivo.
- p62 plays a protective role in various neurodegenerative
disease model flies.
Go to top
Lim, J.Y., Reighard, C.P. and Crowther,
D.C. (2015). The pro-domains of neurotrophins, including
BDNF, are linked to Alzheimer's disease through a toxic synergy
with Aβ. Hum Mol Genet 24: 3929-3938. PubMed ID: 25954034
Abstract
Brain-derived neurotrophic factor (BDNF) has a crucial role in
learning and memory by promoting neuronal survival and modulating
synaptic connectivity. BDNF levels are lower in the brains of
individuals with Alzheimer's disease (AD), suggesting a pathogenic
involvement. The Drosophila orthologue of BDNF is the
highly conserved Neurotrophin 1 (DNT1). BDNF and DNT1 have the
same overall protein structure and can be cleaved, resulting in
the conversion of a full-length polypeptide into separate pro- and
mature-domains. While the BDNF mature-domain is neuroprotective,
the role of the pro-domain is less clear. This study identified a
synergistic toxic interaction between the amyloid-β peptide
(Aβ1–42) and the pro-domains of both DNT1 and BDNF in
flies and mammalian cells. Specifically, it was shown that DNT1
pro-domain acquires a neurotoxic activity in the presence of
Aβ1–42. In contrast, DNT1 mature-domain is protective
against Aβ1–42 toxicity. Likewise, in SH-SY5Y cell
culture, BDNF pro-domain is toxic only in the presence of
Aβ1–42. Western blots indicate that this synergistic
interaction likely results from the Aβ1–42-induced
upregulation of the BDNF pro-domain receptor p75NTR. The clinical
relevance of these findings is underlined by a greater than thirty
fold increase in the ratio of BDNF pro- to mature-domains in the
brains of individuals with AD. This unbalanced BDNF
pro:mature-domain ratio in patients represents a possible
biomarker of AD and may offer a target for therapeutic
intervention (Lim, 2015).
Highlights
- Drosophila DNT1 mRNA levels are high during
development but suppressed in adulthood.
- The subdomains of DNT1 have differential effects on neurotoxic
phenotypes in the fly.
- Co-expression of various domains of DNT1 does not change the
abundance or conformation of Aβ.
- The pro-domain of BDNF and Aβ1–42 exhibit
synergistic toxicity in SH-SY5Y human neuroblastoma cell
cultures.
- An elevated ratio of pro- to mature-domain of BDNF correlates
with disease status in elderly individuals.
Discussion
NTs are a major class of molecules promoting neuronal survival in
vertebrates. They are synthesized as larger precursor forms that
are proteolytically processed to yield a mature, biologically
active ligand. Among the NTs, BDNF has emerged as a major
regulator of synaptic plasticity, neuronal survival and
differentiation, and also as a potential molecular target for the
treatment of neurological disease. Several studies indicate that
the cortex and hippocampus, areas of the brain associated with
learning and memory, not only exhibit extensive amyloid pathology
but also show decreased levels of BDNF in AD. Interestingly,
precursor and mature forms of BDNF are significantly decreased in
preclinical and early stages of AD, and this reduction correlates
with clinical neuropsychological scores. Low levels of BDNF may
favor AD pathogenesis by failing to adequately support neurones
and allowing them to succumb to other toxic insults. Furthermore,
several studies have linked polymorphisms, specifically Val66Met
and Cys270Thr, in the pro-domain of BDNF to an increased risk for
AD (Lim, 2015).
BDNF exerts many of its neuroprotective effects by binding to the
TrkB receptor, a member of the tumor necrosis factor receptor
family. Indeed, some reports indicate that Aβ may have a
negative effect on neuronal survival by down-regulating the TrkB
receptor. For example, levels of TrkB are reduced in the temporal
and frontal cortex of AD brain. In addition, BDNF-induced TrkB
autophosphorylation and the activation of the downstream enzymes
AKT and ERK are all suppressed in the hippocampus of APP/PS1 mice
(Lim, 2015).
The consequences of BDNF binding to its alternative receptor
p75NTR and sortilin are less well understood, although they are
thought to include promotion of myelination and neuronal migration
but also neuronal process retraction and neuronal apoptosis. It
has been proposed that the balance between cell death and survival
is determined by the relative activity of the precursor versus
mature forms of BDNF; indeed, in dorsal root ganglion lesion
models in neonatal rats, the signaling appears to be affected by
the relative levels of the relevant receptors, namely TrkB, p75NTR
and sortilin. Notably, BDNF promotes the death of cultured neurons
in vitro when p75NTR is upregulated and TrkB downregulated.
Precursor BDNF can also cause cell death in both in vitro and in
vivo model systems and the prevailing view is that the apoptotic
signal is generated by the pro-domain. Similar domain-specific
activities have also been observed for NGF; specifically, the
precursor form of this related NT preferentially activates p75NTR
resulting in apoptosis, while mature NGF preferentially activates
TrkA receptor with neurotrophic effects (Lim, 2015).
Mammalian NTs are similar in many ways to their insect
orthologues. Sequence analysis has identified Drosophila
neurotrophin (DNT1), also called Spatzle 2 (Spz 2), as the closest
fly orthologue of human BDNF. Like the NTs, the Spz polypeptides
are synthesized with a signal peptide, followed by a pro-domain
and then the cysteine knot-containing mature domain. The
characteristic NT cysteine knot, formed by antiparallel
β-sheets held together by three disulfide bonds, is conserved
in the crystal structures of both Spz and NGF. There is also
functional conservation between DNT1, Spz and the mammalian NTs in
the nervous system. Indeed, during Drosophila
embryogenesis DNT1 is expressed in neurons and muscle cells where
it promotes neuronal survival and suppresses apoptosis. This study
shows that the high levels of endogenous DNT1 mRNA that
are present during larval development are rapidly suppressed at
the beginning of adult life in both control flies and similarly in
those expressing various isoforms of the Aβ peptide (Lim,
2015).
Because of the functional similarities between mammalian and Drosophila
NTs, this study analyzed the interaction between Aβ and DNT1
in the fruit fly. It was found that transgenic expression of the
DNT1 mature-domain protects flies against Aβ toxicity and
that this benefit is not due to spurious reductions in Aβ
levels. Similarly, in human SH-SY5Y culture, the mature-domain of
BDNF protects cells from Aβ toxicity. In both the fly and the
mammalian models, the pro-domains of both DNT1 and BDNF, while
being harmless alone, are nevertheless toxic when added in
combination with Aβ. In SH-SY5Y cells, this synergistic toxic
interaction between Aβ and the BDNF pro-domain is only
apparent for the Met66 variant and is absent for the wild-type
Val66 isoform. This finding provides mechanistic underpinning for
the clinical observation that the Met66 polymorphism is linked to
poor prognosis, particularly in individuals with high Aβ
burden on PiB photon emission tomography brain scans (Lim, 2015).
The expression of the receptor p75NTR is upregulated in both
SH-SY5Y cells following Aβ treatment and also in transgenic
mice expressing the human APPswe transgene. Aβ treatment also
enhances sortilin expression via p75NTR, which is thought to
activate the downstream effectors JNK and Rho, resulting in
apoptotic cell death. Furthermore, the accumulation of Aβ in
humans is also accompanied by an increased hippocampal
membrane-associated p75NTR. However, it seems unlikely that
Aβ binds directly to p75NTR, rather this study suggests that
Aβ amplifies the pro-apoptotic signaling of pro-domain BDNF
by upregulating p75NTR in the human SH-SY5Y cells (Lim, 2015).
Next, analysis of post-mortem hippocampal tissue from age-matched
healthy elderly and patients with AD underlines the clinical
significance of experimental findings in this study. As expected,
the mature-domain of BDNF is reduced in AD when compared with
control brain tissue. In analysis of 10 cases and controls, it was
found that control subjects fall into two groups—those with
levels of BDNF mature-domain that are up to 20 times higher than
AD cases; however, an equal number of controls have mature-domain
levels that are equivalent to AD patients. The separation between
cases and controls is better when BDNF pro-domain levels are
measured: cases have elevated levels of pro-domain, spread over
almost 2 orders of magnitude while all but one of the controls are
closely grouped. The average pro-domain level is ∼16-fold
higher in AD cases when compared with controls. These reciprocal
changes in the levels of pro- and mature-BDNF are surprising
considering that the peptides are generated stoichiometrically.
Receptor-mediated clearance is an unlikely explanation because the
levels of TrkB are low and p75 high in AD cases when compared with
controls. Conceivably, in AD the BDNF pro-domain is being
stabilized by binding to another protein; this intriguing
possibility requires further investigation (Lim, 2015).
When these relative changes in pro- and mature-domains are
combined as a ratio, it was found that cases and controls have an
average 30-fold difference and only one of the controls overlaps
with the AD range. This particular control individual is
interesting because, despite being symptom-free and relatively
young (aged 63), she was the only control to have accumulated
significant Aβ. In fact, she had the highest level of Aβ
amongst all cases and controls. That this control individual's
pro:mature domain ratio was in the AD range raises the tantalizing
possibility that disease changes can be predicted before the onset
of symptoms (Lim, 2015).
In conclusion, this study shows for the first time that the pro-
and mature-domains of NTs have opposing effects on Aβ
neurotoxicity in vitro and in vivo. The mature-domains of NTs
protect against Aβ1–42 toxicity, whereas the
pro-domains acquir a toxic role in the presence of
Aβ1–42. In case of post-mortem clinical brain samples,
the ratio of pro- to mature-domain of BDNF is significantly higher
in patients with AD when compared with controls. Taken together,
the finding that patients with AD have elevated levels of BDNF
pro-domain underlines the importance of its synergistic toxic
interaction with Aβ. Conceivably, individuals with an
unfavourable pro:mature domain ratio could be targeted with
therapy aimed at restoring a more neurotrophic environment in the
brain (Lim, 2015).
Go to top
Ott, S., Dziadulewicz, N. and Crowther,
D.C. (2015). Iron is a specific cofactor for distinct
oxidation- and aggregation-dependent Aβ toxicity mechanisms
in a Drosophila model. Dis Model Mech 8: 657-667. PubMed
ID: 26035384
Abstract
Metals, including iron, are present at high concentrations in
amyloid plaques in individuals with Alzheimer's disease, where
they are also thought to be cofactors in generating oxidative
stress and modulating amyloid formation. This study presents data
from several Drosophila models of neurodegenerative
proteinopathies indicating that the interaction between iron and
amyloid beta peptide (Aβ) is specific and is not seen for
other aggregation-prone polypeptides. The interaction with iron is
likely to be important in the dimerisation of Aβ and is
mediated by three N-terminal histidines. Transgenic fly lines
systematically expressing all combinations of His>Ala
substitutions in Aβ were generated and used to study the
pathological role of these residues. Developmental eye phenotypes,
longevity and histological examinations indicate that the
N-terminal histidines have distinct position-dependent and
-independent mechanisms. The former mediate the toxic effects of
metals and Aβ aggregation under non-oxidising conditions and
the latter are relevant under oxidising conditions. Understanding
how Aβ mediates neurotoxic effects in vivo will help to
better target pathological pathways using aggregation blockers and
metal-modifying agents (Ott, 2015).
Highlights
- The iron-binding protein ferritin is a specific suppressor of
Aβ toxicity in vivo.
- Iron modifies Aβ aggregation in vitro and in vivo.
- Position-dependent effects of His>Ala substitutions on
Aβ toxicity in a non-oxidising environment.
- Position-dependent effects of His>Ala substitutions on the
metal-dependent component of Aβ toxicity in a non-oxidising
environment.
- Position-independent effects of His>Ala substitutions on
Aβ toxicity in an oxidising environment.
Discussion
A number of neurodegenerative disorders are characterized by the
abnormal metabolism of metals, such as copper and iron; however,
the relevance of this observation to pathological mechanisms
remains unclear. Consequently, this study used several models of
protein aggregation disorders to investigate the role of metals.
In an earlier study using a Drosophila model of Aβ
toxicity, it was found that the iron-binding protein ferritin and
other iron-chelators are powerful suppressors of both Aβ
toxicity and the associated markers of oxidative damage.
Consequently, this study asked whether ferritin expression also
rescues disease-related phenotypes in other Drosophila
models of common neurodegenerative disorders. Specifically, flies
expressing tau,
the Q48 peptide and TDP43,
in addition to various isoforms of Aβ, were analyzed (Ott,
2015).
The inclusion of these particular polypeptides in the study is
justified by varying degrees of evidence linking their
pathogenicity to interactions with metal cofactors. Considering
tau, its hyperphosphorylation and deposition as neurofibrillary
tangles are characteristic of AD and other tauopathies. Some
investigators have highlighted the role of metal binding in
generating tau deposits and the consequent neuronal microtubule
dysfunction. It is thought that this process is induced by the
ability of Fe3+ to bind to phosphorylated tau, causing it to
aggregate. Furthermore, tau, like Aβ, may form complexes with
iron and copper, resulting in the generation of reactive oxygen
species, in turn favouring tau phosphorylation (Ott, 2015).
Similarly, in HD there is evidence for abnormal metal metabolism,
with clinical MRI scans showing iron accumulation in susceptible
brain regions. Iron levels tend to be higher in advanced disease
and in patients with longer polyQ repeat lengths. In mouse and fly
models of HD there is good evidence that both copper and iron
metabolism are disturbed and that chelator therapy may be
beneficial. In vitro, metals promote oxidative damage, with a
truncated huntingtin polypeptide interacting with both Cu2+ and
Fe3+ (Ott, 2015).
Finally, familial amyotrophic
lateral sclerosis (ALS) may result from mutations in the
genes for TDP43, FUS,
C9ORF72 and, classically, SOD1. Although there is considerable
evidence that copper plays a role in the pathogenicity of SOD1
variants, the role of metals in ALS as a whole is less clear. For
example, mice expressing disease-linked variants of TDP43 show
abnormal metabolism of a number of metals, although iron appears
to be normal (Ott, 2015).
Considering this evidence that iron is more or less important in
the pathological actions of a series of aggregation-prone
polypeptides, it is remarkable that, in this study, iron chelation
by ferritin was beneficial only for flies expressing Aβ. The
finding in this study that ferritin offers Aβ-specific rescue
from both longevity and eye phenotypes indicates that an important
component of Aβ toxicity in vivo is not generic. Thus, the
overall toxicity of an aggregating polypeptide seems to be
composed of a core generic component, which is likely to be
related to the presence of oligomeric aggregates. To this core are
added peptide-specific effects that are modulated by environmental
cofactors – in this case the presence of iron or other
metals. One way in which metals could enhance Aβ toxicity is
by stabilising particular conformers in the aggregation pathway.
The formation of an initial dimer is likely to be a fundamental
step and this was investigated in vivo by expressing either the
normal monomeric peptide or a ‘pre-dimerised’ tandem
Aβ peptide. It has been previously shown that both tandem
Aβ42 and tandem Aβ40 aggregate rapidly in vivo; however,
of these, only the 42 amino acid isoform is able to generate
stable oligomeric aggregates and exhibit pronounced toxicity. This
study presents tentative evidence in vitro that amyloid generation
by partially purified, recombinant Aβ and by tandem Aβ
responds differently to the addition of iron. Specifically, the
addition of iron to a preparation of partially purified tandem
Aβ does not cause the slowing of ThT amyloid signal that is
observed with the monomeric peptide preparation. Concordant with
these in vitro results, it was found that the toxicity associated
with tandem Aβ in the fly is not suppressed by co-expression
of ferritin. Furthermore, ferritin has no effect on the number of
tandem Aβ deposits in the brain, whereas flies expressing
monomeric Aβ consistently have up to 25% fewer deposits in
the presence of ferritin. Importantly, this reduced plaque load is
not due to decreased Aβ transgene expression or peptide
production. In summary, these results suggest that, both in vitro
and in vivo, iron is interacting with Aβ to accelerate a step
that is redundant in the tandem peptide; this step is likely to be
the formation of dimeric aggregates. These results in a Drosophila
model are reminiscent of the reduction in plaque load in tg2576
mice treated with the broad-spectrum metal chelator clioquinol and
support the relevance of this study for mammalian systems (Ott,
2015).
After evaluating the specificity of iron for Aβ-induced
toxicity and its effect on aggregation, potential interactions
between Aβ and iron were studied. Such interactions are
thought be mediated by the three N-terminal histidine residues at
positions 6, 13 and 14. The study systematically assessed how each
of the seven possible combinations of His>Ala substitutions
modifies the elaboration of three key disease-linked phenotypes,
namely: (1) survival in an oxidising environment; (2) survival in
a non-oxidising environment; and (3) the proportion of Aβ
toxicity that is metal dependent (Ott, 2015).
To assess the response of the flies to oxidative stress, survival
after oral challenge with hydrogen peroxide was measured. Whereas
control flies are robust when treated in this way, there is a
marked reduction in survival after 78 and 96?h when Aβ is
expressed. Remarkably, only the number of histidines in a
particular Aβ variant determine the susceptibility to
oxidative stress. This histidine dose dependency and position
independence makes it unlikely that the mechanism involves
specific changes in peptide conformation or overall aggregation
propensity; indeed, the hydrogen peroxide feeding experiments are
performed in 6-day-old flies in which there is no detectable
peptide aggregation. Moreover, the predicted aggregation
propensities of the various peptide isoforms are essentially
identical. A possible molecular scale interpretation of these
results is that the formation of redox-active metal-peptide
complexes is rather flexible and peptide promiscuous. In this
model, histidines from distinct peptides could co-ordinate to a
shared metal ion and generate a redox-active complex. An
alternative explanation might be that there is an oxidative
reaction in the presence of Aβ that uses histidines as a
substrate, thereby generating toxic oxygen species. In either
case, these results predict that reducing total brain Aβ
levels might be most effective in reducing the oxidative damage in
AD (Ott, 2015).
By contrast, the remaining two disease-linked phenotypes are
sensitive to the positions of each of the His>Ala
substitutions. In a non-oxidising environment the survival of
flies expressing the various peptides is determined by
position-dependent factors. Some in vitro studies have shown that
the histidines at positions 13 and 14 interact similarly with
copper in models of amyloid fibrils, whereas others suggest that
the H14A substitution in synthetic Aβ42 reduces toxicity,
possibly by preventing the interaction of the peptide with cell
membranes. The longevity effects observed in this study, although
relatively modest, are nevertheless both robust and significant,
and present with a contrasting observation. Specifically, it was
found that H14A enhances toxicity, as evidenced by a reduction in
longevity, whereas the H13A substitution has the opposite effect.
Interestingly, the H6A substitution has a modulating role in the
fly model; in particular, H6A acts to amplify the longevity
effects of substitutions at positions 13 and 14. Accordingly, H14A
reduces the median survival and H6/14A has the shortest lifespan
of all the fly lines. By contrast, Drosophila expressing
the H13A variant live longer than wild-type flies and H6/13A live
the longest of all the peptide-expressing lines. H6 also modulates
the accumulation of peptide deposits in the brain, with H6/14A
showing remarkably high levels of Aβ deposition, higher than
H14A alone. The use of the φC31 system to target all of the
Aβ transgenes to the same 51D genomic insertion site in the
same acceptor fly line makes it highly unlikely that the
phenotypic differences observed in this study are due to
artefactual fluctuations in transgene expression levels.
Furthermore, the genetic background of the experimental flies
should be essentially identical. It should also be noted that the
density of Aβ deposits is comparable between lines despite
highly significant differences in median survival (Ott, 2015).
The modulating role of H6 is seen most clearly in the third
phenotype, where the degree to which the metal chelator clioquinol
prolongs the longevity of the various fly lines was measured. When
the percentage increase in median survival upon treatment with
clioquinol is calculated, it is apparent that the histidines at
positions 13 and 14 each have their position-dependent effects on
the clioquinol response. However, there is an overwhelming
additive effect of histidine at position 6, such that each
corresponding H6A variant shows a 25-30% reduction in clioquinol
responsiveness. The implication of these results is that much of
the metal-mediated toxicity, in a non-oxidising environment, is
dependent on H6A and that, thereafter, there are smaller
position-dependent effects of residues 13 and 14. A possible
mechanistic interpretation is that metal interactions with H6
initiate metal-mediated Aβ dimerisation and, thereafter,
histidines 13 and 14 determine the particular conformations of
downstream aggregates and their toxicity. These position-dependent
properties of individual histidine residues make it likely that
particular peptide conformations are mediating the combined
Aβ-iron effects; such structures might be amenable as
therapeutic targets (Ott, 2015).
In summary, this study's analysis of the effects of metals and
their specific interactions with Aβ in vivo indicates that
dimerisation of the peptide, perhaps mediated by H6, is an early
step in generating toxicity. The subsequent toxic consequences of
Aβ aggregation are then largely determined by the histidines
at positions 13 and 14. In a strongly oxidising environment the
importance of peptide aggregation is complemented, if not
overwhelmed, by the histidine-dependent oxidative damage mediated
by Aβ (Ott, 2015).
Go to top
Sofola-Adesakin, O., Castillo-Quan,
J.I., Rallis, C., Tain, L.S., Bjedov, I., Rogers, I., Li, L.,
Martinez, P., Khericha, M., Cabecinha, M., Bähler, J. and
Partridge, L. (2014). Lithium suppresses Aβ
pathology by inhibiting translation in an adult Drosophila
model of Alzheimer's disease. Front Aging Neurosci 6: 190. PubMed
ID: 25126078
Abstract
The greatest risk factor for Alzheimer's disease (AD) is age, and
changes in the ageing nervous system are likely contributors to AD
pathology. Amyloid beta (Aβ) accumulation, which occurs as a
result of the amyloidogenic processing of amyloid precursor
protein (APP), is thought to initiate the pathogenesis of AD,
eventually leading to neuronal cell death. An adult-onset Drosophila
model of AD has been developed earlier in which mutant Aβ42
accumulation leads to increased mortality and neuronal dysfunction
in the adult flies. Furthermore, lithium reduces Aβ42
protein, but not mRNA, and is able to rescue Aβ42-induced
toxicity. This study investigated the mechanism/s by which lithium
modulates Aβ42 protein levels and Aβ42 induced toxicity
in the fly model. It was found that lithium causes a reduction in
protein synthesis in Drosophila and hence the level of
Aβ42. At both the low and high doses tested, lithium rescues
the locomotory defects induced by Aβ42, but it rescues
lifespan only at lower doses, suggesting that long-term, high-dose
lithium treatment may have induced toxicity. Lithium also
down-regulates translation in the fission yeast Schizosaccharomyces
pombe associated with increased chronological lifespan.
These data highlight a role for lithium and reduced protein
synthesis as potential therapeutic targets for AD pathogenesis
(Sofola-Adesakin, 2014).
Highlights
- Lithium reduces Aβ load in arctic Aβ42 expressing
flies through protein clearance/degradation-independent
mechanisms.
- Lithium down-regulates overall protein synthesis/translation.
- Lithium also inhibits protein synthesis in fission yeast.
- Lithium extended lifespan of flies expressing Aβ.
Discussion
Human life expectancy continues to increase at a steady rate in
most countries worldwide, and has done so by almost 3 months per
year in the last 160 years. Therefore, it is of great importance
to tackle ageing-related diseases such as AD, because they are
becoming increasingly prevalent. Because age is the biggest risk
factor for AD, interventions that promote general increases in
health during ageing could also be important and beneficial in AD
(Sofola-Adesakin, 2014).
Lithium is becoming increasingly implicated as a drug that can
ameliorate ageing and neurodegenerative diseases. Several groups
have shown that it extends lifespan in model organisms such as the
nematode worm C. elegans and Drosophila. This
study shows that lithium also extends lifespan in fission yeast Schizosaccharomyces
pombe, highlighting that this effect is conserved over
large evolutionary distances. Fission yeast is an ideal organism
for genetic screens, and future work should identify the molecular
targets of lithium both for control of protein synthesis and of
lifespan. Furthermore, slightly higher levels of lithium present
in the drinking water have been reported as associated with
reduced mortality in a Japanese human population (Sofola-Adesakin,
2014).
A substantial body of work has demonstrated that several
neurodegenerative diseases and neurological disorders, including
but not confined to stroke, schizophrenia, Fragile
X syndrome, Huntington's
disease and Parkinson's
disease, benefit from the therapeutic properties of lithium.
In addition, several studies have investigated whether lithium has
a beneficial effect in AD pathogenesis. Clinical trials conducted
with lithium have yielded conflicting results; some have found
benefits, whilst others have not. Interestingly, a correlative
study conducted in patients with bipolar disorder, suggests that
patients that have been on chronic lithium treatment show a
reduced incidence of AD in comparison to patients that have not
been on treatment. And a small-scale clinical trial on mild
cognitive impaired (MCI) patients finds that low doses of lithium
slow cognitive decline. The investigators suggest that a reason
for the previous conflicting data on the efficacy of lithium is
probably attributable to the pathological states/stages at which
the patients were given lithium. It is becoming increasingly
evident that drug trials are most likely to yield positive effects
when initiated early, at the MCI stage (Sofola-Adesakin, 2014).
Results from this study add to existing data suggesting that
lithium could be beneficial in ameliorating Aβ toxicity, and
should be considered for a potential large-scale trial on MCI
patients. It has the added advantage of being an already approved
drug, used to treat bipolar patients. It does have side-effects,
but these are minimal at the low doses used in a recent
small-scale clinical study. It was also found that there are
limits to the beneficial/therapeutic benefits of lithium in
fission yeast in chronological lifespan—lithium is unable to
increase chronological lifespan at higher doses as well as in the
Drosophila AD model. Previously, it was shown that
administering both 30 and 100 mM lithium into the fly food is
effective in modulating Aβ neuronal toxicity as evidenced by
the improved locomotor function in young flies. These lithium
concentrations were initially chosen based on another study in
which it was shown that lithium concentrations ranging from 10 to
100 mM lithium in the fly food translates to roughly
0.05–0.4 mM in the fly tissue, so well below the toxic
levels in patients and mice. This study shows that both 25 and 100
mM lithium reduce Aβ levels in a dose dependent manner at an
early time point. It was also found that lower doses of lithium
(10 and 25 mM) rescue the shortened longevity of the Aβ
flies, but 100 mM lithium is unable to extend lifespan when given
to the flies throughout adulthood. It will be important to
determine the therapeutic thresholds for lithium in patients that
could offer therapeutic benefits without overt side effects
(Sofola-Adesakin, 2014).
Similar to the published data on the role of GSK-3 inhibition in
down-regulating translation in HCC1806 cells, it was found that
lithium is able to reduce translation in fission yeast and flies,
suggesting that perhaps some of the effect of lithium on
translation down-regulation is via GSK-3 inhibition. However, this
is correlative and future work will involve carrying out epistasis
interactions between lithium and GSK-3, and identifying molecular
targets of GSK-3 and lithium for control of protein synthesis.
Nonetheless, this study highlights the potential benefits of
lithium through down-regulation of translation, associated with
extension of lifespan in very distantly related organisms. By
reducing protein synthesis, lithium may reduce the increased
proteostatic burden in ageing, a recognized hallmark of ageing.
Lithium is also of specific benefit in AD, because of its ability
to down-regulate translation, and hence levels of proteins
involved in promoting the presence of toxic Aβ
(Sofola-Adesakin, 2014).
The mutant Arctic Aβ42 protein present in the transgenic
flies used in this study has a propensity to aggregate faster than
wild type Aβ. However, both soluble and insoluble Aβ in
the Arctic Aβ42 flies were observed, and the ability of
lithium to reduce translation of the Aβ peptide without
affecting its clearance may lower the level of soluble Aβ. In
a wider context, lithium might be beneficial in ameliorating
toxicity of AD by lowering expression of APP and of proteins that
are involved in the generation of Aβ from APP. The AD model
used in this study does not express full length APP, and may
therefore not include other potential/additional benefits of
lithium on Aβ toxicity. As well as the increased ratio of
Aβ42 to Aβ 40 peptide observed in familial AD cases with
APP mutations, increased levels of APP could also contribute to AD
pathogenesis. Indeed, patients with Down syndrome have a high risk
of developing AD possibly due to trisomy of the APP gene which
leads to increased APP expression. Also, several mutations in the
APP promoter region have been found to significantly increase APP
expression in SH-SY5Y cells, and are associated with risk for AD
(Sofola-Adesakin, 2014).
The ability of lithium to down-regulate translation could
therefore be beneficial at several stages in AD pathogenesis.
Lithium might also have therapeutic benefits for other
neurodegenerative disorders that are caused by over-expression of
wild type or mutant forms of proteins such as α-synuclein in
Parkinson's disease. Lithium could also reduce the production of
mis-translated polypeptides, and free proteases or/and chaperones
that can then participate in cellular proteostasis. Furthermore,
diseases where protein turnover is compromised by loss of function
of the degradation machinery could also benefit from lowering the
burden of protein production hence reducing cellular stress. This
could be particularly important in lysosomal storage diseases,
where the intrinsic function of the degradative machinery is
compromised. Moreover, induction of autophagy in some cases
increases the load of an already dysfunctional lysosome, worsening
the cellular proteostatic stress. Hence lowering the production of
proteins could again be a viable mechanism to restore
proteostasis. Other neurodegenerative models where the role of
lithium in lowering protein translation could be beneficial are,
for example, the Drosophila models of Pink1 and Parkin,
which do not include the over-expression of toxic proteins. Flies
lacking either of these proteins accumulate unfolded proteins in
mitochondria, resulting in mitochondria impairment. It would be
interesting to study whether lithium could ameliorate
mitochondrial stress by reducing the production of the proteins
accumulating in the mitochondria of the Pink1 or parkin
null flies. Lithium could hence be a useful drug with an overall
benefit for health during ageing and protection against AD and
other neurodegenerative diseases (Sofola-Adesakin, 2014).
Go to top
Allan, K., Perez, K.A., Barnham, K.J.,
Camakaris, J. and Burke, R. (2014). A commonly used Drosophila
model of Alzheimer's disease generates an aberrant species of
amyloid-β with an additional N-terminal glutamine residue.
FEBS Lett 588: 3739-3743. PubMed ID: 25171862
Abstract
Expression of human amyloid-β (Aβ) in Drosophila
is frequently used to investigate its toxicity in vivo. This study
expressed Aβ1–42 in the fly using a secretion signal
derived from the Drosophila necrotic
gene, as described in several previous publications.
Surface-enhanced laser desorption/ionization TOF MS analysis
revealed that the Aβ produced contains an additional
glutamine residue at the N-terminus. AβQ+1–42 was found
to have increased protein abundance and to cause more severe
neurodegenerative effects than wild type Aβ1–42 as
assessed by locomotor activity and lifespan assays. These data
reveal that a commonly used model of Alzheimer’s disease
generates incorrect Aβ peptide (Allan, 2014).
Highlights
- Amyloid-β (Aβ) peptide produced with the Necrotic
signal peptide contains an additional N-terminal glutamine.
- Aβ produced with the Necrotic signal peptide gives
dose-dependent neurodegenerative phenotypes.
Discussion
Alzheimer’s disease (AD) is characterized by the presence of
brain extracellular plaques rich in amyloid-β (Aβ), a
peptide derived from the proteolytic cleavage of Amyloid-β
Precursor Protein (AβPP). Differential processing of
AβPP can produce several different Aβ species of varying
lengths. Aβ42 is significantly more cytotoxic than the
abundant Aβ40 normally found in healthy brains and the
Aβ42: Aβ40 ratio is considered to be an important factor
in neuronal health and AD progression (Allan, 2014).
When attempting to replicate AD-like neurodegeneration in the
vinegar fly Drosophila, the need for AβPP
processing has been circumvented by directly expressing Aβ in
fly neuronal tissues. A secretory signal sequence at the
N-terminus of the Aβ peptide, allowing its secretion from
producing cells, is known to be essential for inducing
age-dependent phenotypes including reduced adult lifespan, reduced
locomotor activity, learning defects, deposition of amyloid
plaques and visible brain and retinal degeneration. This model
system has since been used to investigate the effect of single
amino acid substitutions in the Aβ42 peptide sequence and
numerous other aspects of AD including pharmacological and genetic
modification of Aβ-mediated toxicity (Allan, 2014).
This study demonstrates that one of the signal peptides
frequently used to generate secreted Aβ, derived from the fly
Necrotic protein, actually produces an aberrant Aβ1–42
peptide with an additional glutamine residue at the N-terminus,
referred to as AβQ+1–42. A similar situation has been
seen with a C. elegans Aβ transgenic line that
produces a truncated form of Aβ, Aβ3–42, which
begins with glutamic acid. Using transgenic lines with equivalent
transcription levels, it was found that a wild type
Aβ1–42 construct gives dramatically lower peptide
levels and no detectable neurodegenerative phenotypes compared to
the aberrant AβQ+1–42 form (Allan, 2014).
Since TSpAβ1–42 could not be expressed at sufficient
levels for repeating the mass spectrometry analysis performed on
NSpAβ1–42, this study cannot confirm that
TSpAβ1–42 is being correctly processed. However,
Aβ1–42 constructs using the pre-proenkephalin signal
sequence (also predicted to cleave correctly) are toxic when
expressed at high levels. Therefore the study proposes that the
addition of an N-terminal glutamine enhances Aβ abundance and
therefore toxicity compared to wild type Aβ, a property
revealed when the two species are transcribed at similar levels
(Allan, 2014).
The slightly higher NSpAβQ+1–42 transcript levels do
not account for the strong difference in peptide levels seen
between the NSpAβ1–42 and TSpAβ1–42
constructs. Possibly, NSpAβQ+1–42 is more resistant to
degradation and/or clearance. In the human AD brain, N-terminally
truncated Aβ3–42 and Aβ11–42 peptides
represent ∼50% of total Aβ found in senile plaques. Both
peptides begin with a glutamic acid residue which can undergo
post-translational cyclization to produce pyroglutamate (pE).
pE-modified Aβ displays increased stability due to reduced
protease susceptibility. Glutamine can also undergo cyclization.
This study therefore hypothesizes that aberrant signal peptide
cleavage from the NSpAβ1–42 construct results in a
cyclized pQ form of Aβ that displays greater stability and
hence increased toxicity. These findings emphasize the importance
of determining the species of Aβ being generated
experimentally and the transcription levels of each different
construct (Allan, 2014).
Go to top
Hu, Y., Han, Y., Shao, Y., Wang, X.,
Ma, Y., Ling, E. and Xue, L. (2015). Gr33a modulates Drosophila
male courtship preference. Sci Rep 5: 7777. PubMed ID: 25586066
Abstract
In any gamogenetic species, attraction between individuals of the
opposite sex promotes reproductive success that guarantees their
thriving. Consequently, mate determination between two sexes is
effortless for an animal. However, choosing a spouse from numerous
attractive partners of the opposite sex needs deliberation. In Drosophila
melanogaster, both younger virgin females and older ones
are equally liked options to males; nevertheless, when given
options, males prefer younger females to older ones. Non-volatile
cuticular hydrocarbons (CHCs), considered as major pheromones in Drosophila,
constitute females' sexual attraction that act through males'
gustatory receptors (Grs) to elicit male courtship. To date, only
a few putative Grs are known to play roles in male courtship. This
study reports that loss of Gr33a
function or abrogating the activity of Gr33a neurons does not
disrupt male-female courtship, but eliminates males' preference
for younger mates. Furthermore, ectopic expression of human
amyloid precursor protein (APP) in Gr33a neurons abolishes males'
preference behavior. Such function of APP is mediated by the
transcription factor forkhead box O (dFoxO). These results not
only provide mechanistic insights into Drosophila male
courtship preference, but also establish a novel Drosophila
model for Alzheimer's disease (AD) (Hu, 2015).
Highlights
- The roles of Grs in males' courtship preference behavior.
- Gr33a is required for males' preference for younger
mates.
- Activity of Gr33a neurons is essential for males' preference
for younger mates.
- Expression of APP in Gr33a neurons results in dFoxO-dependent
choice defect.
- Female CHC profiles change with age.
Discussion
Drosophila male courtship choice has been frequently
applied for studying decision making in animals, yet most of the
past studies have focused on male courtship choices between likes
and dislikes, such as court towards females vs. males, or virgin
vs. non-virgin females. The choice behavior between two
equally-liked options: mature virgin females, whether younger or
older, has been previously shown to be similarly attractive to
naive males; nevertheless, when given the option, males turn out
to be picky and prefer younger virgin females to older ones. This
study shows that a gustatory receptor, Gr33a, is necessary for
males' preference for younger mates. Gr33a is thought to be
necessary to inhibit homosexual behavior; its role in heterosexual
behavior, however, is rarely pondered. This study reveals the
critical role of Gr33a in males' preference for younger mates.
Furthermore, ectopic expression of APP in Gr33a neurons eliminates
males' preference behavior, and such function is mediated by
dFoxO, a recently reported downstream factor of APP. Therefore,
this study demonstrates the genetic interaction of APP and dFoxO
in Gr33a neurons, which modulates males' preference for younger
mates (Hu, 2015).
APP is identified as a potential causative protein of AD, a
common progressive neurodegenerative disorder, in which cognitive
decline is the prime symptom. Although Drosophila has
long been utilized for building AD models to investigate the
pathogenesis and possible cure for AD, accepted Drosophila
AD models are limited to locomotion model and life span model,
which have little correlation with cognitive ability. Findings in
this study, however, offer the possibility for establishing a
novel Drosophila AD model that is related to cognitive
ability (Hu, 2015).
Go to top
Lau, H.C., Lee, I.K., Ko, P.W., Lee,
H.W., Huh, J.S., Cho, W.J. and Lim, J.O. (2015).
Non-invasive screening for Alzheimer's disease by sensing salivary
sugar using Drosophila cells expressing gustatory
receptor (Gr5a) immobilized on an extended gate ion-sensitive
field-effect transistor (EG-ISFET) biosensor. PLoS One 10:
e0117810. PubMed ID: 25714733
Abstract
Body fluids are often used as specimens for medical diagnosis.
With the advent of advanced analytical techniques in
biotechnology, the diagnostic potential of saliva has been the
focus of many studies. A previous study reports the presence of
excess salivary sugars, in patients with Alzheimer’s disease
(AD). This study developed a highly sensitive, cell-based
biosensor to detect trehalose levels in patient saliva. The
developed biosensor relies on the overexpression of sugar
sensitive gustatory receptors (Gr5a)
in Drosophila cells to detect the salivary trehalose.
The cell-based biosensor is built on the foundation of an improved
extended gate ion-sensitive field-effect transistor (EG-ISFET).
Using an EG-ISFET, instead of a traditional ion-sensitive
field-effect transistor (ISFET), results in an increase in the
sensitivity and reliability of detection. The biosensor is
designed with the gate terminals segregated from the conventional
ISFET device. This design allows the construction of an
independent reference and sensing region for simultaneous and
accurate measurements of samples from controls and patients
respectively. To investigate the efficacy of the cell-based
biosensor for AD screening, 20 saliva samples were collected from
each of the following groups: participants diagnosed with AD,
participants diagnosed with Parkinson’s disease (PD), and a
control group composed of healthy individuals. Then, the response
generated from the interaction of the salivary trehalose of the
saliva samples and the Gr5a in the immobilized cells on an
EG-ISFET sensor was studies. The cell-based biosensor
significantly distinguishes salivary sugar, trehalose of the AD
group from the PD and control groups. Based on these findings, the
study propose that salivary trehalose might be a potential
biomarker for AD and could be detected using the cell-based
EG-ISFET biosensor developed in this study. The cell-based
EG-ISFET biosensor provides a sensitive and direct approach for
salivary sugar detection and may be used in the future as a
screening method for AD (Lau, 2015).
Go to top
Frenkel-Pinter, M., Tal, S., Scherzer-Attali, R., Abu-Hussien, M., Alyagor, I., Eisenbaum, T., Gazit, E. and Segal, D. (2016). Naphthoquinone-tryptophan hybrid inhibits aggregation of the Tau-derived peptide PHF6 and reduces neurotoxicity. J Alzheimers Dis [Epub ahead of print]. PubMed ID: 26836184
Abstract
Tauopathies, such as Alzheimer's disease (AD), are a group of disorders characterized neuropathologically by intracellular toxic accumulations of abnormal protein aggregates formed by misfolding of the microtubule-associated protein tau. Since protein self-assembly appears to be an initial key step in the pathology of this group of diseases, intervening in this process can be both a prophylactic measure and a means for modifying the course of the disease for therapeutic purposes. Aromatic small molecules can be effective inhibitors of aggregation of various protein assemblies, by binding to the aromatic core in aggregation-prone motifs and preventing their self-assembly. Specifically, series of small aromatic naphthoquinone-tryptophan hybrid molecules were designed as candidate aggregation inhibitors of β-sheet based assembly, and their efficacy was demonstrated toward inhibiting aggregation of the amyloid-β peptide, another culprit of AD, as well as of various other aggregative proteins involved in other protein misfolding diseases. This study tested whether a leading naphthoquinone-tryptophan hybrid molecule, namely NQTrp, can be repurposed as an inhibitor of the aggregation of the tau protein in vitro and in vivo. The molecule was shown to inhibit the in vitro assembly of PHF6, the aggregation-prone fragment of tau protein, reduces hyperphosphorylated tau deposits and ameliorates tauopathy-related behavioral defect in an established transgenic Drosophila model expressing human tau. It is suggested that NQTrp, or optimized versions of it, could act as novel disease modifying drugs for AD and other tauopathies (Frenkel-Pinter, 2016).
Go to top
Bouge, A. L. and Parmentier, M. L. (2016). Tau excess impairs mitosis and kinesin-5 function, leading to aneuploidy and cell death. Dis Model Mech [Epub ahead of print]. PubMed ID: 26822478
Abstract
In neurodegenerative diseases like Alzheimer's disease (AD), cell cycle defects and associated aneuploidy have been described. However, the importance of these defects in the physiopathology of AD and the underlying mechanistic processes are largely unknown in particular with respect to the microtubule-binding protein Tau, which is found in excess in the brain and cerebrospinal fluid of patients. Although it has long been known that Tau is phosphorylated during mitosis to generate a lower affinity for microtubules, there has been no indication that an excess of this protein could affect mitosis. The effect of an excess of human Tau (hTau) protein on cell mitosis was studied in vivo. Using the Drosophila developing wing disc epithelium as a model, this study shows that an excess of hTau induces a mitotic arrest, with the presence of monopolar spindles. This mitotic defect leads to aneuploidy and apoptotic cell death. The mechanism of action of hTau was studied and it was found that the MT-binding domain of hTau is responsible for these defects. hTau effects occur via the inhibition of the function of the kinesin Klp61F, the Drosophila homologue of kinesin-5 (also called Eg5 or KIF11). This deleterious effect of hTau is also found in other Drosophila cell types (neuroblasts) and tissues (the developing eye disc) as well as in human Hela cells.By demonstrating that microtubule-bound Tau inhibits the Eg5/KIF11 kinesin and cell mitosis, this work provides a new framework to consider the role of Tau in neurodegenerative diseases (Bouge, 2016).
Go to top
Kilian, J.G., Hsu, H.W., Mata, K., Wolf, F.W.
and Kitazawa, M. (2017). Astrocyte
transport of glutamate and neuronal activity reciprocally modulate tau
pathology in Drosophila. Neuroscience [Epub ahead of
print]. PubMed ID: 28215745
Abstract
Abnormal buildup of the microtubule associated protein tau is a major
pathological hallmark of Alzheimer's
disease (AD) and various tauopathies. The mechanisms by which
pathological tau accumulates and spreads throughout the brain remain
largely unknown. It is known that a restoration of the major astrocytic
glutamate transporter, GLT1, ameliorates a buildup of tau pathology and
rescues cognition in a mouse model of AD. In this study, it was
hypothesized that aberrant extracellular glutamate and abnormal neuronal
excitatory activities promote tau
pathology. Consequently, the genetic interactions between tau and the GLT1
homolog dEaat1 were investigated in
Drosophila melanogaster. Neuronal-specific overexpression of
human wildtype tau markedly shortens lifespan and impairs motor behavior.
RNAi depletion of dEaat1 in astrocytes worsens these phenotypes, whereas
overexpression of dEaat1 improves them. However, the synaptic neuropil
appears unaffected, and there is no major neuronal loss with tau
overexpression in combination with dEaat1 depletion. To mimic
glutamate-induced aberrant excitatory input in neurons, repeated
depolarization of neurons via transgenic TrpA1
was applied to the adult Drosophila optic nerves, and the change
of tau deposits was examined. Repeated depolarization significantly
increases the accumulation of tau in these neurons. The study propose that
increased neuronal excitatory activity exacerbates tau-mediated neuronal
toxicity and behavioral deficits (Killian, 2017).
Go to top
Li, A., Hooli, B., Mullin, K., Tate, R.E.,
Bubnys, A., Kirchner, R., Chapman, B., Hofmann, O., Hide, W. and Tanzi,
R.E. (2017).
Silencing of the Drosophila
ortholog of Sox5 leads to abnormal neuronal development and behavioral
impairment.
Hum Mol Genet [Epub ahead of print]. PubMed ID: 28186563
Abstract
SOX5 encodes
a transcription factor that is expressed in multiple tissues including
heart, lung and brain. Mutations in SOX5 have been previously
found in patients with amyotrophic
lateral sclerosis (ALS) and developmental delay, intellectual
disability and dysmorphic features. To characterize the neuronal role of SOX5,
this study silenced the Drosophila ortholog of SOX5, Sox102F,
by RNAi in various neuronal subtypes in Drosophila. Silencing of
Sox102F leads to misorientated and disorganized michrochaetes,
neurons with shorter dendritic arborization (DA) and reduced complexity,
diminished larval peristaltic contractions, loss of neuromuscular
junction bouton structures, impaired olfactory
perception, and severe neurodegeneration in brain. Silencing of SOX5
in human SH-SY5Y neuroblastoma cells results in a significant repression
of WNT signaling activity and altered expression of WNT-related genes.
Samples of SOX5 variants reveals several variants that
show significant association with Alzheimers disease disease status. These findings indicate that SOX5
is a novel candidate gene for AD with important role in neuronal
function. The genetic findings warrant further studies to identify and
characterize SOX5 variants that confer risk for AD, ALS and
intellectual disability.
Go to top
Cutler, T., Sarkar, A., Moran, M.,
Steffensmeier, A., Puli, O.R., Mancini, G., Tare, M., Gogia, N.
and Singh, A. (2015). Drosophila eye model to
study neuroprotective role of CREB Binding Protein (CBP) in
Alzheimer's disease. PLoS One 10: e0137691. PubMed ID: 25714733
Abstract
The progressive neurodegenerative disorder Alzheimer's disease
(AD) manifests as loss of cognitive functions, and finally leads
to death of the affected individual. AD may result from
accumulation of amyloid plaques. These amyloid plaques comprising
of amyloid-beta 42 (Aβ42) polypeptides results from the
improper cleavage of amyloid precursor protein (APP) in the brain.
The Aβ42 plaques have been shown to disrupt the normal
cellular processes and thereby trigger abnormal signaling which
results in the death of neurons. However, the molecular-genetic
mechanism(s) responsible for Aβ42 mediated neurodegeneration
is yet to be fully understood. This study utilized Gal4/UAS system
to develop a transgenic fruit fly model for Aβ42 mediated
neurodegeneration. Targeted misexpression of human Aβ42 in
the differentiating photoreceptor neurons of the developing eye of
transgenic fly triggers neurodegeneration. This progressive
neurodegenerative phenotype resembles Alzheimer's like
neuropathology. The histone acetylase, CREB Binding Protein (CBP),
was identified as a genetic modifier of Aβ42 mediated
neurodegeneration. Targeted misexpression of CBP along with
Aβ42 in the differentiating retina can significantly rescue
neurodegeneration. It was found that gain-of-function of CBP
rescues Aβ42 mediated neurodegeneration by blocking cell
death. Misexpression of Aβ42 affects the targeting of axons
from retina to the brain but misexpression of full length CBP
along with Aβ42 can restore this defect. The CBP protein has
multiple domains and is known to interact with many different
proteins. Structure-function analysis using truncated constructs
lacking one or more domains of CBP protein in transgenic flies
reveals that Bromo, HAT and polyglutamine (BHQ) domains together
are required for the neuroprotective function of CBP. This BHQ
domain of CBP has not been attributed to promote survival in any
other neurodegenerative disorders. In conclusion, the study
identifies CBP as a genetic modifier of Aβ42 mediated
neurodegeneration. Furthermore, BHQ domain of CBP is responsible
for its neuroprotective function (Cutler, 2015).
Go to top
Jonson, M., Pokrzywa, M., Starkenberg,
A., Hammarstrom, P. and Thor, S. (2015). Systematic
Aβ analysis in Drosophila reveals high toxicity for
the 1-42, 3-42 and 11-42 peptides, and emphasizes N- and
C-terminal residues. PLoS One 10: e0133272. PubMed ID: 25714733
Abstract
Brain amyloid plaques are a hallmark of Alzheimer's disease (AD),
and primarily consist of aggregated Aβ peptides. While
Aβ 1-40 and Aβ 1-42 are the most abundant, a number of
other Aβ peptides have also been identified. Studies have
indicated differential toxicity for these various Aβ
peptides, but in vivo toxicity has not been systematically tested.
To address this issue, this study generated improved transgenic Drosophila
UAS strains expressing 11 pertinent Aβ peptides. UAS
transgenic flies were generated by identical chromosomal
insertion, hence removing any transgenic position effects, and
crossed to a novel and robust Gal4 driver line. Using this
improved Gal4/UAS set-up, survival and activity assays revealed
that Aβ 1-42 severely shortens lifespan and reduces activity.
N-terminal truncated peptides are quite toxic, with 3-42 similar
to 1-42, while 11-42 shows a pronounced but less severe phenotype.
N-terminal mutations in 3-42 (E3A) or 11-42 (E11A) result in
reduced toxicity for 11-42, and reduced aggregation for both
variants. Strikingly, C-terminal truncation of Aβ (1-41, -40,
-39, -38, -37) are non-toxic. In contrast, C-terminal extension to
1-43 results in reduced lifespan and activity, but not to the same
extent as 1-42. Mutating residue 42 in 1-42 (A42D, A42R and A42W)
greatly reduces Aβ accumulation and toxicity. Histological
and biochemical analysis reveals strong correlation between in
vivo toxicity and brain Aβ aggregate load, as well as amount
of insoluble Aβ. This systematic Drosophila in vivo
and in vitro analysis reveals crucial N- and C-terminal
specificity for Aβ neurotoxicity and aggregation, and
underscores the importance of residues 1-10 and E11, as well as a
pivotal role of A42 (Jonson, 2015).
Go to top
Haddadi, M., Nongthomba, U., Jahromi, S. R. and Ramesh, S. R. (2015). Transgenic Drosophila model to study apolipoprotein E4-induced neurodegeneration. Behav Brain Res [Epub ahead of print]. PubMed ID: 26706888
Abstract
The ε4 isoform of Apolipoprotein E (ApoE4) that is involved in neuron-glial lipid metabolism has been demonstrated as the main genetic risk factor in late-onset of Alzheimer's disease. However, the mechanism underlying ApoE4-mediated neurodegeneration remains unclear. This study created a transgenic model of neurodegenerative disorder by expressing ε3 and ε4 isoforms of human ApoE in the Drosophila. The genetic models exhibited progressive neurodegeneration, shortened lifespan and memory impairment. Genetic interaction studies between amyloid precursor protein and ApoE in axon pathology of the disease revealed that over expression of hApoE in Appl-expressing neurons of Drosophila brain causes neurodegeneration. This Drosophila model may facilitate analysis of the molecular and cellular events implicated in hApoE4 neurotoxicity.
Go to top
Ali, Y. O., Ruan, K. and Zhai, R. G.
(2012). NMNAT suppresses tau-induced neurodegeneration by promoting clearance of hyperphosphorylated tau oligomers in a Drosophila model of tauopathy. Hum Mol Genet 21(2): 237-250. PubMed ID: 21965302