Proline-rich transmembrane protein 2 (PRRT2) is the causative gene for a heterogeneous group of familial paroxysmal neurological disorders that include seizures with onset in the first year of life (benign familial infantile seizures), paroxysmal kinesigenic dyskinesia or a combination of both. Most of the PRRT2 mutations are loss-of-function leading to haploinsufficiency and 80% of the patients carry the same frameshift mutation (c.649dupC; p.Arg217Profs*8), which leads to a premature stop codon. To model the disease and dissect the physiological role of PRRT2, we studied the phenotype of neurons differentiated from induced pluripotent stem cells from previously described heterozygous and homozygous siblings carrying the c.649dupC mutation. Single- cell patch-clamp experiments on induced pluripotent stem cell-derived neurons from homozygous patients showed increased Na + currents that were fully rescued by expression of wild-type PRRT2. Closely similar electrophysiological features were observed in primary neurons obtained from the recently characterized PRRT2 knockout mouse. This phenotype was associated with an increased length of the axon initial segment and with markedly augmented spontaneous and evoked firing and bursting activities evaluated, at the network level, by multi-electrode array electrophysiology. Using HEK-293 cells stably expressing Na v channel subtypes, we demonstrated that the expression of PRRT2 decreases the membrane exposure and Na + current of Na v 1.2/Na v 1.6, but not Na v 1.1, channels. Moreover, PRRT2 directly interacted with Na v 1.2/Na v 1.6 channels and induced a negative shift in the voltage-dependence of inactivation and a slow-down in the recovery from inactivation. In addition, by co-immunoprecipitation assays, we showed that the PRRT2-Na v interaction also occurs in brain tissue. The study demonstrates that the lack of PRRT2 leads to a hyperactivity of voltage-dependent Na + channels in homozygous PRRT2 knockout human and mouse neurons and that, in addition to the reported synaptic functions, PRRT2 is an important negative modulator of Na v 1.2 and Na v 1.6 channels. Given the predominant paroxysmal character of PRRT2-linked diseases, the disturbance in cellular excitability by lack of negative modulation of Na + channels appears as the key pathogenetic mechanism.

PRRT2 controls neuronal excitability by negatively modulating Na + channel 1.2/1.6 activity / F. Fruscione, P. Valente, B. Sterlini, A. Romei, S. Baldassari, M. Fadda, C. Prestigio, G. Giansante, J. Sartorelli, P. Rossi, A. Rubio, A. Gambardella, T. Nieus, V. Broccoli, A. Fassio, P. Baldelli, A. Corradi, F. Zara, F. Benfenati. - In: BRAIN. - ISSN 0006-8950. - 141:4(2018 Apr 01), pp. 1000-1016. [10.1093/brain/awy051]

PRRT2 controls neuronal excitability by negatively modulating Na + channel 1.2/1.6 activity

T. Nieus;
2018

Abstract

Proline-rich transmembrane protein 2 (PRRT2) is the causative gene for a heterogeneous group of familial paroxysmal neurological disorders that include seizures with onset in the first year of life (benign familial infantile seizures), paroxysmal kinesigenic dyskinesia or a combination of both. Most of the PRRT2 mutations are loss-of-function leading to haploinsufficiency and 80% of the patients carry the same frameshift mutation (c.649dupC; p.Arg217Profs*8), which leads to a premature stop codon. To model the disease and dissect the physiological role of PRRT2, we studied the phenotype of neurons differentiated from induced pluripotent stem cells from previously described heterozygous and homozygous siblings carrying the c.649dupC mutation. Single- cell patch-clamp experiments on induced pluripotent stem cell-derived neurons from homozygous patients showed increased Na + currents that were fully rescued by expression of wild-type PRRT2. Closely similar electrophysiological features were observed in primary neurons obtained from the recently characterized PRRT2 knockout mouse. This phenotype was associated with an increased length of the axon initial segment and with markedly augmented spontaneous and evoked firing and bursting activities evaluated, at the network level, by multi-electrode array electrophysiology. Using HEK-293 cells stably expressing Na v channel subtypes, we demonstrated that the expression of PRRT2 decreases the membrane exposure and Na + current of Na v 1.2/Na v 1.6, but not Na v 1.1, channels. Moreover, PRRT2 directly interacted with Na v 1.2/Na v 1.6 channels and induced a negative shift in the voltage-dependence of inactivation and a slow-down in the recovery from inactivation. In addition, by co-immunoprecipitation assays, we showed that the PRRT2-Na v interaction also occurs in brain tissue. The study demonstrates that the lack of PRRT2 leads to a hyperactivity of voltage-dependent Na + channels in homozygous PRRT2 knockout human and mouse neurons and that, in addition to the reported synaptic functions, PRRT2 is an important negative modulator of Na v 1.2 and Na v 1.6 channels. Given the predominant paroxysmal character of PRRT2-linked diseases, the disturbance in cellular excitability by lack of negative modulation of Na + channels appears as the key pathogenetic mechanism.
proline-rich transmembrane protein 2; paroxysmal disorders; induced pluripotent stem cells; voltage-dependent sodium channels; neuronal excitability
Settore BIO/09 - Fisiologia
1-apr-2018
Article (author)
File in questo prodotto:
File Dimensione Formato  
Fruscione et al. 2018

accesso aperto

Tipologia: Publisher's version/PDF
Dimensione 2.25 MB
Formato Adobe PDF
2.25 MB Adobe PDF Visualizza/Apri
Pubblicazioni consigliate

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/2434/568339
Citazioni
  • ???jsp.display-item.citation.pmc??? 34
  • Scopus 90
  • ???jsp.display-item.citation.isi??? 87
social impact