Abstract

Proteinopathy in neurodegenerative diseases is typically characterized by deteriorating activity of specific protein aggregates. In tauopathies, including Alzheimer’s disease (AD), tau protein abnormally accumulates and induces dysfunction of the affected neurons. Despite active identification of tau modifications responsible for tau aggregation, a critical modulator inducing tau proteinopathy by affecting its protein degradation flux is not known. Here, we report that anaplastic lymphoma kinase (ALK), a receptor tyrosine kinase, is crucial for the tau-mediated AD pathology. ALK caused abnormal accumulation of highly phosphorylated tau in the somatodendritic region of neurons through its tyrosine kinase activity. ALK-induced LC3-positive axon swelling and loss of spine density, leading to tau-dependent neuronal degeneration. Notably, ALK activation in neurons impaired Stx17-dependent autophagosome maturation and this defect was reversed by a dominant-negative Grb2. In a Drosophila melanogaster model, transgenic flies neuronally expressing active Drosophila Alk exhibited the aggravated tau rough eye phenotype with retinal degeneration and shortened lifespan. In contrast, expression of kinase-dead Alk blocked these phenotypes. Consistent with the previous RNAseq analysis showing upregulation of ALK expression in AD [1], ALK levels were significantly elevated in the brains of AD patients showing autophagosomal defects. Injection of an ALK.Fc-lentivirus exacerbated memory impairment in 3xTg-AD mice. Conversely, pharmacologic inhibition of ALK activity with inhibitors reversed the memory impairment and tau accumulation in both 3xTg-AD and tauC3 (caspase-cleaved tau) transgenic mice. Together, we propose that aberrantly activated ALK is a bona fide mediator of tau proteinopathy that disrupts autophagosome maturation and causes tau accumulation and aggregation, leading to neuronal dysfunction in AD.

Highlights

  • These authors contributed : Jisu Park, Hyunwoo Choi

  • Likewise, treating primary hippocampal neurons with mAb46, an agonistic anaplastic lymphoma kinase (ALK) antibody [40], increased tau levels and its phosphorylation, while pre-treatment with mAb30, an antagonistic ALK antibody [40], blocked mAb46-mediated tau accumulation (Fig. 1D). We confirmed that both mAb46 and mAb30 antibodies well detected mouse ALK in the transfected cells (Fig. S1d)

  • We found that ALK phosphorylated human and mouse tau at multiple serine and threonine residues, including the Alzheimer’s disease (AD) epitopes PHF-1 (S396/ S404), 12E8 (S262/S356), CP13 (S202) and pThr231‐tau (Figs. 1B–D and S1g)

Read more

Summary

1234567890();,: 1234567890();,: Introduction

Aberrant ALK activity has been strongly implicated in the oncogenesis of human cancer as a fusion protein in inflammatory myofibroblastic tumors, diffuse large B-cell lymphoma and anaplastic large-cell lymphoma, or through mutations in the full-length protein in hereditary familial neuroblastoma [18,19,20,21,22]. This makes ALK a therapeutic target in cancer [23,24,25]. Our results define a crucial role of ALK in tau-mediated neurodegeneration and provide insight into the pathogenesis of AD and a new approach to AD therapeutics

Materials and methods
Discussion
Compliance with ethical standards
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call