Abstract

The microtubule-associated protein tau has a critical role in Alzheimer’s disease and other tauopathies. A proposed pathomechanism in the progression of tauopathies is the trans-synaptic spreading of tau seeds, with a role for exosomes which are secretory nanovesicles generated by late endosomes. Our previous work demonstrated that brain-derived exosomes isolated from tau transgenic rTg4510 mice encapsulate tau seeds with the ability to induce tau aggregation in recipient cells. We had also shown that exosomes can hijack the endosomal pathway to spread through interconnected neurons. Here, we reveal how tau seeds contained within internalized exosomes exploit mechanisms of lysosomal degradation to escape the endosome and induce tau aggregation in the cytosol of HEK293T-derived ‘tau biosensor cells’. We found that the majority of the exosome-containing endosomes fused with lysosomes to form endolysosomes. Exosomes induced their permeabilization, irrespective of the presence of tau seeds, or whether the exosomal preparations originated from mouse brains or HEK293T cells. We also found that permeabilization is a conserved mechanism, operating in both non-neuronal tau biosensor cells and primary neurons. However, permeabilization of endolysosomes only occurred in a small fraction of cells, which supports the notion that permeabilization occurs by a thresholded mechanism. Interestingly, tau aggregation was only induced in cells that exhibited permeabilization, presenting this as an escape route of exosomal tau seeds into the cytosol. Overexpression of RAB7, which is required for the formation of endolysosomes, strongly increased tau aggregation. Conversely, inhibition of lysosomal function with alkalinizing agents, or by knocking-down RAB7, decreased tau aggregation. Together, we conclude that the enzymatic activities of lysosomes permeabilize exosomal and endosomal membranes, thereby facilitating access of exosomal tau seeds to cytosolic tau to induce its aggregation. Our data underscore the importance of endosomal membrane integrity in mechanisms of cellular invasion by misfolded proteins that are resistant to lysosomal degradation.

Highlights

  • Neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), frontotemporal lobar degeneration with tau (FTLD-tau) and amyotrophic lateral sclerosis (ALS) are proteinopathies, characterized by the misfolding and aggregation of signature proteins [58]

  • We first addressed whether lysosomes are colocalized with internalized exogenous exosomes which had been isolated from P301L tau transgenic rTg4510 and wild-type brains [59, 60]

  • Error bars represent ± SD for 29 individual cells analyzed from four independent experiments. ****p < 0.0001; ns, not significant. o Colocalization, in that M1 represents the proportion of exosomal signal that colocalizes with the Lysotracker signal, and M2 the fraction of the Lysotracker signal colocalizing with the exosomal fluorophore in tau biosensor cells treated with wild-type (Wt) or rTg4510-derived (Tg) exosomes. p–s Increasing the cellular pH of tau biosensor cells with the alkalinizing agent ammonium chloride ­(NH4Cl) reduces tau aggregation induced by exosomes: (p–r top panels) Epifluorescence microscopy detecting tau RD-YFP in cells before flow cytometry

Read more

Summary

Introduction

Neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), frontotemporal lobar degeneration with tau (FTLD-tau) and amyotrophic lateral sclerosis (ALS) are proteinopathies, characterized by the misfolding and aggregation of signature proteins [58]. Acta Neuropathologica (2021) 141:235–256 its pattern of spreading led to the proposition that AD progression may occur by neuron-to-neuron transmission via trans-synaptic transport of misfolded tau seeds from affected to anatomically connected neurons [11, 12]. It is generally believed that a prion-like mechanism is adopted, meaning that misfolded tau seeds actively corrupt the proper folding of soluble tau in recipient cells [51]. Trans-neuronal transfer of tau seeds can be achieved by several mechanisms that involve extracellular vesicles such as exosomes or microvesicles [21, 60, 77], tunneling nanotubes that establish a direct connection between the cytoplasm of neighboring cells [69], or trans-synaptic transfer of membrane-free tau seeds between interconnected neurons [16, 22]

Methods
Results
Discussion
Conclusion
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