Helmholtz Gemeinschaft

Search
Browse
Statistics
Feeds

A CRISPR-Cas9-engineered mouse model for GPI-anchor deficiency mirrors human phenotypes and exhibits hippocampal synaptic dysfunctions

[img]
Preview
PDF (Original Article) - Requires a PDF viewer such as GSview, Xpdf or Adobe Acrobat Reader
4MB
[img] Other (Supporting Information)
104MB

Item Type:Article
Title:A CRISPR-Cas9-engineered mouse model for GPI-anchor deficiency mirrors human phenotypes and exhibits hippocampal synaptic dysfunctions
Creators Name:Rodríguez de los Santos, M. and Rivalan, M. and David, F.S. and Stumpf, A. and Pitsch, J. and Tsortouktzidis, D. and Velasquez, L.M. and Voigt, A. and Schilling, K. and Mattei, D. and Long, M. and Vogt, G. and Knaus, A. and Fischer-Zirnsak, B. and Wittler, L. and Timmermann, B. and Robinson, P.N. and Horn, D. and Mundlos, S. and Kornak, U. and Becker, A.J. and Schmitz, D. and Winter, Y. and Krawitz, P.M.
Abstract:Pathogenic germline mutations in PIGV lead to glycosylphosphatidylinositol biosynthesis deficiency (GPIBD). Individuals with pathogenic biallelic mutations in genes of the glycosylphosphatidylinositol (GPI)-anchor pathway exhibit cognitive impairments, motor delay, and often epilepsy. Thus far, the pathophysiology underlying the disease remains unclear, and suitable rodent models that mirror all symptoms observed in human patients have not been available. Therefore, we used CRISPR-Cas9 to introduce the most prevalent hypomorphic missense mutation in European patients, Pigv:c.1022C > A (p.A341E), at a site that is conserved in mice. Mirroring the human pathology, mutant Pigv(341E) mice exhibited deficits in motor coordination, cognitive impairments, and alterations in sociability and sleep patterns, as well as increased seizure susceptibility. Furthermore, immunohistochemistry revealed reduced synaptophysin immunoreactivity in Pigv(341E) mice, and electrophysiology recordings showed decreased hippocampal synaptic transmission that could underlie impaired memory formation. In single-cell RNA sequencing, Pigv(341E)-hippocampal cells exhibited changes in gene expression, most prominently in a subtype of microglia and subicular neurons. A significant reduction in Abl1 transcript levels in several cell clusters suggested a link to the signaling pathway of GPI-anchored ephrins. We also observed elevated levels of Hdc transcripts, which might affect histamine metabolism with consequences for circadian rhythm. This mouse model will not only open the doors to further investigation into the pathophysiology of GPIBD, but will also deepen our understanding of the role of GPI-anchor–related pathways in brain development.
Keywords:GPI-Anchor Deficiency, Disease Modeling, Hippocampal Synaptic Defect, Animals, Mice
Source:Proceedings of the National Academy of Sciences of the United States of America
ISSN:0027-8424
Publisher:National Academy of Sciences
Volume:118
Number:2
Page Range:e2014481118
Date:12 January 2021
Official Publication:https://doi.org/10.1073/pnas.2014481118
PubMed:View item in PubMed

Repository Staff Only: item control page

Downloads

Downloads per month over past year

Open Access
MDC Library