PUBLICATION

Autolysosome biogenesis and developmental senescence are regulated by both Spns1 and v-ATPase

Authors
Sasaki, T., Lian, S., Khan, A., Llop, J.R., Samuelson, A.V., Chen, W., Klionsky, D.J., Kishi, S.
ID
ZDB-PUB-161123-2
Date
2017
Source
Autophagy   13(2): 386-403 (Journal)
Registered Authors
Chen, Wenbiao, Kishi, Shuji
Keywords
Aging, autophagy, lysosome, senescence, spinster, vacuolar-type H+-ATPase, zebrafish
MeSH Terms
  • Animals
  • Autophagy*
  • Base Sequence
  • Biomarkers/metabolism
  • CRISPR-Cas Systems/genetics
  • Cellular Senescence*
  • Embryo, Nonmammalian/cytology
  • Embryo, Nonmammalian/metabolism
  • Gene Expression Regulation, Developmental
  • Gene Knockdown Techniques
  • Longevity
  • Lysosomes/metabolism*
  • Membrane Fusion
  • Membrane Proteins/metabolism*
  • Phagosomes/metabolism
  • Proton-Translocating ATPases/metabolism*
  • Survival Analysis
  • Vacuolar Proton-Translocating ATPases/metabolism*
  • Zebrafish/embryology*
  • Zebrafish/genetics
  • Zebrafish Proteins/metabolism*
PubMed
27875093 Full text @ Autophagy
Abstract
Spns1 (Spinster homolog 1 [Drosophila]) in vertebrates, as well as Spin (Spinster) in Drosophila, is a hypothetical lysosomal H+-carbohydrate transporter, which functions at a late stage of macroautophagy (hereafter autophagy). The Spin/Spns1 defect induces aberrant autolysosome formation that leads to developmental senescence in the embryonic stage and premature aging symptoms in adulthood. However, the molecular mechanism by which loss of Spin/Spns1 leads to the specific pathogenesis remains to be elucidated. Using chemical, genetic and CRISPR/Cas9-mediated genome-editing approaches in zebrafish, we investigated and determined a mechanism that suppresses embryonic senescence as well as autolysosomal impairment mediated by Spns1 deficiency. Unexpectedly, we found that a concurrent disruption of the vacuolar-type H+-ATPase (v-ATPase) subunit gene, atp6v0ca (ATPase, H+ transporting, lysosomal, V0 subunit ca) led to suppression of the senescence induced by the Spns1 defect, whereas the sole loss of Atp6v0ca led to senescent embryos similar to the single spns1 mutation. Moreover, we discovered that the combined stable defect seen in the presence of both the spns1 and atp6v0ca mutant genes still subsequently induced premature autophagosome-lysosome fusion marked by insufficient acidity, while extending developmental life span, compared with the solely mutated spns1 defect. Our data suggest that Spns1 and the v-ATPase orchestrate proper autolysosomal biogenesis with optimal acidification that is critically linked to developmental senescence and survival.
Genes / Markers
Figures
Show all Figures
Expression
Phenotype
Mutations / Transgenics
Human Disease / Model
Sequence Targeting Reagents
Fish
Antibodies
Orthology
Engineered Foreign Genes
Mapping