Abstract

ATP6V1B2 encodes the V1B2 subunit in V-ATPase, a proton pump responsible for the acidification of lysosomes. Mutations in this gene cause DDOD syndrome, DOORS syndrome, and Zimmermann–Laband syndrome, which share overlapping feature of congenital sensorineural deafness, onychodystrophy, and different extents of intellectual disability without or with epilepsy. However, the underlying mechanisms remain unclear. To investigate the pathological role of mutant ATP6V1B2 in the auditory system, we evaluated auditory brainstem response, distortion product otoacoustic emissions, in a transgenic line of mice carrying c.1516 C > T (p.Arg506∗) in Atp6v1b2, Atp6v1b2Arg506*/Arg506*. To explore the pathogenic mechanism of neurodegeneration in the auditory pathway, immunostaining, western blotting, and RNAscope analyses were performed in Atp6v1b2Arg506*/Arg506* mice. The Atp6v1b2Arg506*/Arg506* mice showed hidden hearing loss (HHL) at early stages and developed late-onset hearing loss. We observed increased transcription of Atp6v1b1 in hair cells of Atp6v1b2Arg506*/Arg506* mice and inferred that Atp6v1b1 compensated for the Atp6v1b2 dysfunction by increasing its own transcription level. Genetic compensation in hair cells explains the milder hearing impairment in Atp6v1b2Arg506*/Arg506* mice. Apoptosis activated by lysosomal dysfunction and the subsequent blockade of autophagic flux induced the degeneration of spiral ganglion neurons and further impaired the hearing. Intraperitoneal administration of the apoptosis inhibitor, BIP-V5, improved both phenotypical and pathological outcomes in two live mutant mice. Based on the pathogenesis underlying hearing loss in Atp6v1b2-related syndromes, systemic drug administration to inhibit apoptosis might be an option for restoring the function of spiral ganglion neurons and promoting hearing, which provides a direction for future treatment.

Highlights

  • V-ATPase is a multi-subunit enzyme complex known as the vacuolar H+-ATPase

  • DPOAE threshold of 4–32 kHz showed no significant difference between Atp6v1b2Arg506∗/Arg506∗ (4 kHz: 41.67 ± 3.33, 8 kHz: 38.33 ± 1.67, 16 kHz: 43.33 ± 3.33, 32 kHz: 65 ± 5.77, n = 3) and WT mice (4 kHz: 40 ± 2.89, 8 kHz: 38.33 ± 4.41, 16 kHz: 45 ± 5.77, 32 kHz: 68.33 ± 3.33, n = 3) (T-test, P > 0.05, Supplementary Figure 1D) at 6 months, which indicated the normal function of outer hair cells (OHCs)

  • The results showed that in the cochlea of Atp6v1b2Arg506∗/Arg506∗ mice, apoptosis was mostly observed in the spiral ganglia (Figure 3E), and no excessive apoptosis was identified in the organ of Corti (Supplementary Figure 4C)

Read more

Summary

Introduction

V-ATPase is a multi-subunit enzyme complex known as the vacuolar H+-ATPase This proton pump is mainly responsible for the acidification of lysosomes and other membrane-bound compartments (Beyenbach and Wieczorek, 2006). The interaction between the V1B2 and V1E subunits was found to be weaker in Atp6v1b2Arg506∗/Arg506∗ mice than in wild-type (WT) mice, indicating that the assembly of V-ATPase was affected by the mutation (Zhao et al, 2019). In another prior study, when pIRES2-EGFP-ATP6V1B2 WT and pIRES2EGFP-ATP6V1B2 c.1516. Abnormalities in autophagy caused by V-ATPase defects are associated with neurodegenerative diseases (Peric and Annaert, 2015; Cerri and Blandini, 2019)

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