Pompe disease results in a Golgi-based glycosylation deficit in human induced pluripotent stem cell-derived cardiomyocytes.

TitlePompe disease results in a Golgi-based glycosylation deficit in human induced pluripotent stem cell-derived cardiomyocytes.
Publication TypeJournal Article
Year of Publication2015
AuthorsRaval KK, Tao R, White BE, De Lange WJ, Koonce CH, Yu J, Kishnani PS, Thomson JA, Mosher DF, Ralphe JC, Kamp TJ
JournalJ Biol Chem
Volume290
Issue5
Pagination3121-36
Date Published2015 Jan 30
ISSN1083-351X
KeywordsBlotting, Western, Cells, Cultured, Genotype, Glycogen Storage Disease Type II, Glycosylation, Golgi Apparatus, Humans, Immunohistochemistry, Induced Pluripotent Stem Cells, Myocytes, Cardiac
Abstract

Infantile-onset Pompe disease is an autosomal recessive disorder caused by the complete loss of lysosomal glycogen-hydrolyzing enzyme acid α-glucosidase (GAA) activity, which results in lysosomal glycogen accumulation and prominent cardiac and skeletal muscle pathology. The mechanism by which loss of GAA activity causes cardiomyopathy is poorly understood. We reprogrammed fibroblasts from patients with infantile-onset Pompe disease to generate induced pluripotent stem (iPS) cells that were differentiated to cardiomyocytes (iPSC-CM). Pompe iPSC-CMs had undetectable GAA activity and pathognomonic glycogen-filled lysosomes. Nonetheless, Pompe and control iPSC-CMs exhibited comparable contractile properties in engineered cardiac tissue. Impaired autophagy has been implicated in Pompe skeletal muscle; however, control and Pompe iPSC-CMs had comparable clearance rates of LC3-II-detected autophagosomes. Unexpectedly, the lysosome-associated membrane proteins, LAMP1 and LAMP2, from Pompe iPSC-CMs demonstrated higher electrophoretic mobility compared with control iPSC-CMs. Brefeldin A induced disruption of the Golgi in control iPSC-CMs reproduced the higher mobility forms of the LAMPs, suggesting that Pompe iPSC-CMs produce LAMPs lacking appropriate glycosylation. Isoelectric focusing studies revealed that LAMP2 has a more alkaline pI in Pompe compared with control iPSC-CMs due largely to hyposialylation. MALDI-TOF-MS analysis of N-linked glycans demonstrated reduced diversity of multiantennary structures and the major presence of a trimannose complex glycan precursor in Pompe iPSC-CMs. These data suggest that Pompe cardiomyopathy has a glycan processing abnormality and thus shares features with hypertrophic cardiomyopathies observed in the congenital disorders of glycosylation.

DOI10.1074/jbc.M114.628628
Alternate JournalJ. Biol. Chem.
PubMed ID25488666
PubMed Central IDPMC4317045
Grant ListP41 GM103490 / GM / NIGMS NIH HHS / United States
P41GM10349010 / GM / NIGMS NIH HHS / United States
R01 HL107367 / HL / NHLBI NIH HHS / United States
T32 GM008692 / GM / NIGMS NIH HHS / United States
U01 HL099773 / HL / NHLBI NIH HHS / United States
U01 HL099773 / HL / NHLBI NIH HHS / United States