How mutant prion protein (PrP) leads to neurological dysfunction in genetic prion diseases is unknown. Tg(PG14) mice synthesize a misfolded mutant PrP which is partially retained in the neuronal endoplasmic reticulum (ER). As these mice age, they develop ataxia and massive degeneration of cerebellar granule neurons (CGNs). Here, we report that motor behavioral deficits in Tg(PG14) mice emerge before neurodegeneration and are associated with defective glutamate exocytosis from granule neurons due to impaired calcium dynamics. We found that mutant PrP interacts with the voltage-gated calcium channel α 2δ-1 subunit, which promotes the anterograde trafficking of the channel. Owing to ER retention of mutant PrP, α 2δ-1 accumulates intracellularly, impairing delivery of the channel complex to the cell surface. Thus, mutant PrP disrupts cerebellar glutamatergic neurotransmission by reducing the number of functional channels in CGNs. These results link intracellular PrP retention to synaptic dysfunction, indicating new modalities of neurotoxicity and potential therapeutic strategies. How mutant prion protein causes neurological dysfunction in genetic prion diseases is not fully known. Senatore et al. report intracellular accumulation of misfolded prion impairs voltage-gated calcium channel transport to synapses, altering glutamatergic neurotransmission and cerebellum-dependent motor function.

Mutant PrP suppresses glutamatergic neurotransmission in cerebellar granule neurons by impairing membrane delivery of VGCC α 2δ-1 subunit

Matteoli M;
2012-01-01

Abstract

How mutant prion protein (PrP) leads to neurological dysfunction in genetic prion diseases is unknown. Tg(PG14) mice synthesize a misfolded mutant PrP which is partially retained in the neuronal endoplasmic reticulum (ER). As these mice age, they develop ataxia and massive degeneration of cerebellar granule neurons (CGNs). Here, we report that motor behavioral deficits in Tg(PG14) mice emerge before neurodegeneration and are associated with defective glutamate exocytosis from granule neurons due to impaired calcium dynamics. We found that mutant PrP interacts with the voltage-gated calcium channel α 2δ-1 subunit, which promotes the anterograde trafficking of the channel. Owing to ER retention of mutant PrP, α 2δ-1 accumulates intracellularly, impairing delivery of the channel complex to the cell surface. Thus, mutant PrP disrupts cerebellar glutamatergic neurotransmission by reducing the number of functional channels in CGNs. These results link intracellular PrP retention to synaptic dysfunction, indicating new modalities of neurotoxicity and potential therapeutic strategies. How mutant prion protein causes neurological dysfunction in genetic prion diseases is not fully known. Senatore et al. report intracellular accumulation of misfolded prion impairs voltage-gated calcium channel transport to synapses, altering glutamatergic neurotransmission and cerebellum-dependent motor function.
2012
Age Factors; Animals; Newborn; Biophysics; Calcium; Calcium Channels; Cells; Cultured; Cerebellum; Disease Models; Animal; Electric Stimulation; Endoplasmic Reticulum; Glutamic Acid; Green Fluorescent Proteins; Magnetic Resonance Imaging; Membrane Potentials; Mice; Inbred C57BL; Transgenic; Motor Activity; Mutation; Neurons; Patch-Clamp Techniques; Prion Diseases; Prions; Protein Transport; Reaction Time; Rotarod Performance Test; Synaptic Transmission; Synaptosomes; Neuroscience (all)
File in questo prodotto:
Non ci sono file associati a questo prodotto.

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/11699/6670
Citazioni
  • ???jsp.display-item.citation.pmc??? 38
  • Scopus 56
  • ???jsp.display-item.citation.isi??? 50
social impact