Congenital disorders of glycosylation (CDG) comprise a family of human multisystemic diseases caused by recessive mutations in genes required for protein N-glycosylation. More than 100 distinct forms of CDGs have been identified and most of them cause severe neurological impairment. The Conserved Oligomeric Golgi (COG) complexmediates tethering of vesicles carrying glycosylation enzymes across the Golgi cisternae. Mutations affecting human COG1, COG2 and COG4-COG8 cause monogenic forms of inherited, autosomal recessive CDGs.We have generated a Drosophila COG7-CDG model that closely parallels the pathological characteristics of COG7-CDG patients, including pronounced neuromotor defects associated with altered N-glycome profiles. Consistent with these alterations, larval neuromuscular junctions of Cog7 mutants exhibit a significant reduction in bouton numbers. We demonstrate that the COG complex cooperates with Rab1 and Golgi phosphoprotein 3 to regulate Golgi trafficking and that overexpression of Rab1 can rescue the cytokinesis and locomotor defects associated with loss of Cog7. Our results suggest that the Drosophila COG7-CDG model can be used to test novel potential therapeutic strategies by modulating trafficking pathways.

Frappaolo, A., Sechi, S., Kumagai, T., Robinson, S., Fraschini, R., Ghahnavieh, A., et al. (2017). COG7 deficiency in Drosophila generates multifaceted developmental, behavioral and protein glycosylation phenotypes. JOURNAL OF CELL SCIENCE, 130(21), 3637-3649 [10.1242/jcs.209049].

COG7 deficiency in Drosophila generates multifaceted developmental, behavioral and protein glycosylation phenotypes

Fraschini, R;
2017

Abstract

Congenital disorders of glycosylation (CDG) comprise a family of human multisystemic diseases caused by recessive mutations in genes required for protein N-glycosylation. More than 100 distinct forms of CDGs have been identified and most of them cause severe neurological impairment. The Conserved Oligomeric Golgi (COG) complexmediates tethering of vesicles carrying glycosylation enzymes across the Golgi cisternae. Mutations affecting human COG1, COG2 and COG4-COG8 cause monogenic forms of inherited, autosomal recessive CDGs.We have generated a Drosophila COG7-CDG model that closely parallels the pathological characteristics of COG7-CDG patients, including pronounced neuromotor defects associated with altered N-glycome profiles. Consistent with these alterations, larval neuromuscular junctions of Cog7 mutants exhibit a significant reduction in bouton numbers. We demonstrate that the COG complex cooperates with Rab1 and Golgi phosphoprotein 3 to regulate Golgi trafficking and that overexpression of Rab1 can rescue the cytokinesis and locomotor defects associated with loss of Cog7. Our results suggest that the Drosophila COG7-CDG model can be used to test novel potential therapeutic strategies by modulating trafficking pathways.
Articolo in rivista - Articolo scientifico
COG7; Drosophila; GOLPH3; Glycosylation; Golgi
English
2017
130
21
3637
3649
partially_open
Frappaolo, A., Sechi, S., Kumagai, T., Robinson, S., Fraschini, R., Ghahnavieh, A., et al. (2017). COG7 deficiency in Drosophila generates multifaceted developmental, behavioral and protein glycosylation phenotypes. JOURNAL OF CELL SCIENCE, 130(21), 3637-3649 [10.1242/jcs.209049].
File in questo prodotto:
File Dimensione Formato  
10281-173293.pdf

accesso aperto

Tipologia di allegato: Publisher’s Version (Version of Record, VoR)
Dimensione 6.42 MB
Formato Adobe PDF
6.42 MB Adobe PDF Visualizza/Apri
sechi et al 2017.pdf

Solo gestori archivio

Tipologia di allegato: Publisher’s Version (Version of Record, VoR)
Dimensione 2.9 MB
Formato Adobe PDF
2.9 MB Adobe PDF   Visualizza/Apri   Richiedi una copia

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/10281/173293
Citazioni
  • Scopus 20
  • ???jsp.display-item.citation.isi??? 21
Social impact