ESPE Abstracts (2018) 89 FC1.6

aWilliam Harvey Research Institute, Centre for Endocrinology, Barts and the London School of Medicine, Queen Mary University of London, London, UK; bWellcome Trust Sanger Institute, Cambridge, UK; cWilliam Harvey Research Institute, Centre for Translational Bioinformatics, Barts and the London School of Medicine, Queen Mary University of London, London, UK; 4The Jackson Laboratory for Genomic Medicine, Farmington, CT USA


Triple A syndrome (AAAS) is a rare, incurable, recessive disorder, characterised by achalasia, alacrima, adrenal failure and a neurodegenerative phenotype. The AAAS gene encodes ALADIN, is a nuclear pore complex (NPC) protein necessary for nuclear import of DNA protective molecules, important for redox homeostasis. ALADIN’s role is not fully characterised: its discovery at the centrosome and the endoplasmic reticulum suggests a role outside the NPC. To date, the interrogation of ALADIN’s function has been limited by suboptimal disease models: knock-out mice do not exhibit an analogous phenotype, patient fibroblast cultures do not originate from the affected tissue-type, and knock-down experiments with immortalised cell lines achieve at best an 80% reduction in ALADIN expression.

Aim: To generate cellular models of AAAS with isogenic controls and undertake characterisation.

Method: We have developed induced pluripotent stem cell (iPSC) models of AAAS using CRISPR-Cas9 gene-editing: (i) Bi-allelic exon 2 deletion (AAAS-KO) and (ii) AAAS homozygous patient mutation: a splice donor hotspot mutation p.G14fs (c.43C>A, exon 1) (AAAS-mutant). These are paired with the original healthy wild-type (WT) iPSC line and mono-allelic exon 2 deletion (AAAS-het) as isogenic controls.

Results: Immunoblotting did not detect ALADIN in AAAS-KO or AAAS-mutant cells. There was no difference in cellular proliferation after 24 hours between AAAS-KO compared to WT by cell counting (P value 0.24). Immunofluorescence with Ki67 antibody confirmed there were no significant changes in cellular proliferation (WT: 100% of cells exhibit Ki67 compared to 88.46% of AAAS-KO cells, P value 0.40). RNA sequencing was performed to identify differences in the transcriptome of the iPSC lines, comparing WT to AAAS-KO, AAAS-mutant and a mono-allelic exon 2 deletion (AAAS-het) (n=3). This identified 9 genes with significantly altered transcription (LogFC values >1.1 and <−1.1) with preliminary analysis suggesting an impact of AAAS deficiency on genes involved in oxidative stress. We demonstrated that AAAS-KO and AAAS-mutant cells will differentiate along a neurocortical lineage, expressing neuronal transcription factors OTX2 PAX6 and Nestin.

Conclusion: We present a viable alternative iPSC model for the study of ALADIN in a near endogenous environment. These can be differentiated along a neurocortical lineage, to reflect the tissue affected in the Triple A Syndrome. We present detailed transcriptome analysis, which will inform further functional experiments, designed to provide insight into the pathogenesis of AAAS.

Article tools

My recent searches

No recent searches.

My recently viewed abstracts