4.6 Article

Uncoupling neuronal death and dysfunction in Drosophila models of neurodegenerative disease

Journal

ACTA NEUROPATHOLOGICA COMMUNICATIONS
Volume 4, Issue -, Pages -

Publisher

BMC
DOI: 10.1186/s40478-016-0333-4

Keywords

Alzheimer disease; Parkinson disease; MAPT; Tau; Amyloid-beta peptide; SNCA; alpha-synuclein; Neurodegeneration; Neurophysiology; Synapses; Animal model; Drosophila

Categories

Funding

  1. NIH [K08AG034290, R21NS089854, R01AG033193, U01AG046161, R01AG050631, C06RR029965, P01ES16732, P30CA125123]
  2. Alzheimer's Association
  3. American Federation for Aging Research
  4. Huffington Foundation
  5. Robert and Renee Belfer Family Foundation
  6. Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital
  7. Career Award for Medical Scientists from the Burroughs Wellcome Fund
  8. Eunice Kennedy Shriver National Institute of Child Health & Human Development [U54HD083092]

Ask authors/readers for more resources

Common neurodegenerative proteinopathies, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are characterized by the misfolding and aggregation of toxic protein species, including the amyloid beta (A beta) peptide, microtubule-associated protein Tau (Tau), and alpha-synuclein (alpha Syn) protein. These factors also show toxicity in Drosophila; however, potential limitations of prior studies include poor discrimination between effects on the adult versus developing nervous system and neuronal versus glial cell types. In addition, variable expression paradigms and outcomes hinder systematic comparison of toxicity profiles. Using standardized conditions and medium-throughput assays, we express human Tau, A beta or alpha Syn selectively in neurons of the adult Drosophila retina and monitor age-dependent changes in both structure and function, based on tissue histology and recordings of the electroretinogram (ERG), respectively. We find that each protein causes a unique profile of neurodegenerative pathology, demonstrating distinct and separable impacts on neuronal death and dysfunction. Strikingly, expression of Tau leads to progressive loss of ERG responses whereas retinal architecture and neuronal numbers are largely preserved. By contrast, A beta induces modest, age-dependent neuronal loss without degrading the retinal ERG. alpha Syn expression, using a codon-optimized transgene, is characterized by marked retinal vacuolar change, progressive photoreceptor cell death, and delayed-onset but modest ERG changes. Lastly, to address potential mechanisms, we perform transmission electron microscopy (TEM) to reveal potential degenerative changes at the ultrastructural level. Surprisingly, Tau and alpha Syn each cause prominent but distinct synaptotoxic profiles, including disorganization or enlargement of photoreceptor terminals, respectively. Our findings highlight variable and dynamic properties of neurodegeneration triggered by these disease-relevant proteins in vivo, and suggest that Drosophila may be useful for revealing determinants of neuronal dysfunction that precede cell loss, including synaptic changes, in the adult nervous system.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available