2015
DOI: 10.1038/mt.2015.106
|View full text |Cite
|
Sign up to set email alerts
|

Arylsulfatase A Overexpressing Human iPSC-derived Neural Cells Reduce CNS Sulfatide Storage in a Mouse Model of Metachromatic Leukodystrophy

Abstract: Metachromatic leukodystrophy (MLD) is an inherited lysosomal storage disorder resulting from a functional deficiency of arylsulfatase A (ARSA), an enzyme that catalyzes desulfation of 3-O-sulfogalactosylceramide (sulfatide). Lack of active ARSA leads to the accumulation of sulfatide in oligodendrocytes, Schwann cells and some neurons and triggers progressive demyelination, the neuropathological hallmark of MLD. Several therapeutic approaches have been explored, including enzyme replacement, autologous hematopo… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
1
1
1
1

Citation Types

2
29
0

Year Published

2016
2016
2022
2022

Publication Types

Select...
7
1
1

Relationship

1
8

Authors

Journals

citations
Cited by 44 publications
(31 citation statements)
references
References 40 publications
(35 reference statements)
2
29
0
Order By: Relevance
“…Intracerebral cell grafts of murine neural precursors overexpressing arylsulfatase A resulted in sulfatide reduction and modest improvement of a murine ARSA-deficient model (Givogri et al, 2006; Kawabata et al, 2006). Similar results were subsequently obtained using neural progenitors derived from ESCs and iPSCs (Doerr et al, 2015; Klein et al, 2006). Although the donor cells were capable of in vivo oligodendroglial and astroglial differentiation, they did so at low frequency, possibly accounting for the relatively modest treatment-associated suppression of demyelination with disease progression.…”
Section: Disease Targets For Neural and Glial Progenitor Cell Therapysupporting
confidence: 75%
“…Intracerebral cell grafts of murine neural precursors overexpressing arylsulfatase A resulted in sulfatide reduction and modest improvement of a murine ARSA-deficient model (Givogri et al, 2006; Kawabata et al, 2006). Similar results were subsequently obtained using neural progenitors derived from ESCs and iPSCs (Doerr et al, 2015; Klein et al, 2006). Although the donor cells were capable of in vivo oligodendroglial and astroglial differentiation, they did so at low frequency, possibly accounting for the relatively modest treatment-associated suppression of demyelination with disease progression.…”
Section: Disease Targets For Neural and Glial Progenitor Cell Therapysupporting
confidence: 75%
“…Lt-NES cells represent a stable neural stem cell population, which can be extensively propagated while maintaining a stable neurogenic potential yielding fully functional neurons both in vitro and following transplantation into the rodent brain1617. Due to their robust proliferation and differentiation potential, lt-NES cells lend themselves particularly well to genetic modification and have been successfully used for lineage tracing, disease modelling and cell-mediated gene transfer181920. They exhibit a posterior phenotype with an anterior hindbrain identity, and mostly give rise to GABAergic interneurons as well as glutamatergic neurons, a differentiation pattern maintained after transplantation into neonatal and adult hosts1621.…”
Section: Resultsmentioning
confidence: 99%
“…To facilitate fast enrichment of lentivirally transduced cells, the expression cassette was transferred to the pLVXTP backbone which contains a puromycin resistance gene (Clonetech) by opening it using XhoI and MluI digestion and ligation with the PCR amplified (forward primer 5′-GCAGCAGTCGACACTGCAGAGGGCCCTGC-3′; reverse primer 5′-GCAGCAACGCGTTTGTCTGAGGTGTGACTGGAAAACC-3′) and SalI / MluI digested tracing cassette. Lentiviral particles were generated and concentrated using the polyethylene glycol method as described previously2043. Briefly, 293FT cells (Life Technologies) were transfected with the lentiviral plasmids pLVX-SynHTB or pLentiWE-Ef1α-mRFP1 and the packaging plasmid psPAX2 and the envelope plasmid pMD2.G.…”
Section: Methodsmentioning
confidence: 99%
“…Our RNAseq data revealed that the G9a mutant labyrinth overexpresses secreted protein-encoding genes, including secreted modular calcium-binding protein 1 (Smoc1) and arylsulfatase family member 1 (Arsi). Smoc1 is expressed in endothelial cells and positively regulates cell proliferation (Awwad et al, 2015), and Arsi is secreted from proliferating long-term self-renewing neuroepithelial-like stem cells (Doerr et al, 2015). Thus, placental endothelial cells might limit trophoblast proliferation via G9a-mediated repression of secreted activators of cell proliferation.…”
Section: Discussionmentioning
confidence: 99%