Abstract

During prion infections of the central nervous system (CNS) the cellular prion protein, PrPC, is templated to a conformationally distinct form, PrPSc. Recent studies have demonstrated that the Sprn gene encodes a GPI-linked glycoprotein Shadoo (Sho), which localizes to a similar membrane environment as PrPC and is reduced in the brains of rodents with terminal prion disease. Here, analyses of prion-infected mice revealed that down-regulation of Sho protein was not related to Sprn mRNA abundance at any stage in prion infection. Down-regulation was robust upon propagation of a variety of prion strains in Prnp a and Prnp b mice, with the exception of the mouse-adapted BSE strain 301 V. In addition, Sho encoded by a TgSprn transgene was down-regulated to the same extent as endogenous Sho. Reduced Sho levels were not seen in a tauopathy, in chemically induced spongiform degeneration or in transgenic mice expressing the extracellular ADan amyloid peptide of familial Danish dementia. Insofar as prion-infected Prnp hemizygous mice exhibited accumulation of PrPSc and down-regulation of Sho hundreds of days prior to onset of neurologic symptoms, Sho depletion can be excluded as an important trigger for clinical disease or as a simple consequence of neuronal damage. These studies instead define a disease-specific effect, and we hypothesize that membrane-associated Sho comprises a bystander substrate for processes degrading PrPSc. Thus, while protease-resistant PrP detected by in vitro digestion allows post mortem diagnosis, decreased levels of endogenous Sho may trace an early response to PrPSc accumulation that operates in the CNS in vivo. This cellular response may offer new insights into the homeostatic mechanisms involved in detection and clearance of the misfolded proteins that drive prion disease pathogenesis.

Highlights

  • Prion diseases, including the prototypical scrapie of sheep and Creutzfeldt-Jakob Disease (CJD) of humans, are fatal and incurable neurodegenerative disorders

  • As prioninfected mice with only one copy of the PrP gene exhibited both accumulation of PrPSc and a reduction of Sho protein hundreds of days prior to onset of neurologic symptoms, the drop in Sho protein level can be excluded as an important trigger for clinical disease, or a nonspecific consequence of brain cell damage

  • Our studies define a effect restricted to prion disease and we hypothesize that Sho protein is a ‘‘bystander’’ for degradative processes aimed at destroying PrPSc

Read more

Summary

Introduction

Prion diseases, including the prototypical scrapie of sheep and Creutzfeldt-Jakob Disease (CJD) of humans, are fatal and incurable neurodegenerative disorders. They are unusual in that they are often transmissible or infectious diseases. In the disease process a benign, host-encoded a-helical glycoprotein (prion protein, PrPC) undergoes a conformational transition to a b-sheet enriched and infectivity-associated form commonly denoted PrPSc (sometimes denoted as PrPd). This transition is often marked by reduced detergent solubility and acquisition of resistance to proteinase K (PK) digestion in vitro. In strong and independent support of a central role for altered PrP in disease pathogenesis and replication of the transmissible agent, missense mutations in the murine prion

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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