[HTML][HTML] Pathological modeling of glycogen storage disease type III with CRISPR/Cas9 edited human pluripotent stem cells

L Rossiaud, P Fragner, E Barbon, A Gardin… - Frontiers in Cell and …, 2023 - frontiersin.org
L Rossiaud, P Fragner, E Barbon, A Gardin, M Benabides, E Pellier, J Cosette, L El Kassar…
Frontiers in Cell and Developmental Biology, 2023frontiersin.org
Introduction: Glycogen storage disease type III (GSDIII) is a rare genetic disease caused by
mutations in the AGL gene encoding the glycogen debranching enzyme (GDE). The
deficiency of this enzyme, involved in cytosolic glycogen degradation, leads to pathological
glycogen accumulation in liver, skeletal muscles and heart. Although the disease manifests
with hypoglycemia and liver metabolism impairment, the progressive myopathy is the major
disease burden in adult GSDIII patients, without any curative treatment currently available …
Introduction: Glycogen storage disease type III (GSDIII) is a rare genetic disease caused by mutations in the AGL gene encoding the glycogen debranching enzyme (GDE). The deficiency of this enzyme, involved in cytosolic glycogen degradation, leads to pathological glycogen accumulation in liver, skeletal muscles and heart. Although the disease manifests with hypoglycemia and liver metabolism impairment, the progressive myopathy is the major disease burden in adult GSDIII patients, without any curative treatment currently available.
Methods: Here, we combined the self-renewal and differentiation capabilities of human induced pluripotent stem cells (hiPSCs) with cutting edge CRISPR/Cas9 gene editing technology to establish a stable AGL knockout cell line and to explore glycogen metabolism in GSDIII.
Results: Following skeletal muscle cells differentiation of the edited and control hiPSC lines, our study reports that the insertion of a frameshift mutation in AGL gene results in the loss of GDE expression and persistent glycogen accumulation under glucose starvation conditions. Phenotypically, we demonstrated that the edited skeletal muscle cells faithfully recapitulate the phenotype of differentiated skeletal muscle cells of hiPSCs derived from a GSDIII patient. We also demonstrated that treatment with recombinant AAV vectors expressing the human GDE cleared the accumulated glycogen.
Discussion: This study describes the first skeletal muscle cell model of GSDIII derived from hiPSCs and establishes a platform to study the mechanisms that contribute to muscle impairments in GSDIII and to assess the therapeutic potential of pharmacological inducers of glycogen degradation or gene therapy approaches.
Frontiers