Abstract

The pathological cascade leading from primary storage to neural cell dysfunction and death in metachromatic leukodystrophy (MLD) has been poorly elucidated in human-derived neural cell systems. In the present study, we have modeled the progression of pathological events during the differentiation of patient-specific iPSCs to neuroepithelial progenitor cells (iPSC-NPCs) and mature neurons, astrocytes, and oligodendrocytes at the morphological, molecular, and biochemical level. We showed significant sulfatide accumulation and altered sulfatide composition during the differentiation of MLD iPSC-NPCs into neuronal and glial cells. Changes in sulfatide levels and composition were accompanied by the expansion of the lysosomal compartment, oxidative stress, and apoptosis. The neuronal and glial differentiation capacity of MLD iPSC-NPCs was significantly impaired. We showed delayed appearance and/or reduced levels of oligodendroglial and astroglial markers as well as reduced number of neurons and disorganized neuronal network. Restoration of a functional Arylsulfatase A (ARSA) enzyme in MLD cells using lentiviral-mediated gene transfer normalized sulfatide levels and composition, globally rescuing the pathological phenotype. Our study points to MLD iPSC-derived neural progeny as a useful in vitro model to assess the impact of ARSA deficiency along NPC differentiation into neurons and glial cells. In addition, iPSC-derived neural cultures allowed testing the impact of ARSA reconstitution/overexpression on disease correction and, importantly, on the biology and functional features of human NPCs, with important therapeutic implications.

Highlights

  • Introduction MetachromaticLeukodystrophy (MLD) is a rare genetic lysosomal storage disorder (LSD) caused by the functional deficiency of Arylsulfatase A (ARSA; EC 3.1.6.8)

  • Human induced pluripotent stem cells (iPSCs)-derived cultures highly enriched in neurons are used to model the neuronal defects of mucopolysaccharidosis[42,43,44], Niemann−Pick type C45–47, Gaucher disease[48,49,50,51], GM1 gangliosidosis[52], and NCL53

  • The neural conversion of iPSCs triggered the acquisition of a neural-specific sulfatide composition[20] and promoted sulfatide storage in ARSA-deficient neuroepithelial progenitor cells (NPCs)

Read more

Summary

Introduction

Introduction MetachromaticLeukodystrophy (MLD) is a rare genetic lysosomal storage disorder (LSD) caused by the functional deficiency of Arylsulfatase A (ARSA; EC 3.1.6.8). A Histogram plots showing the percentages of cells expressing markers of NPCs (NESTIN) and glial precursors (A2B5, NG2), oligodendrocytes (OLIG2, O4, CNPase, APC), astroglial (GFAP), and neuronal cells (β-tubulin III) at d0, d14, and d24 of differentiation in ND, MLD, and MLD-ARSA cultures.

Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.