Helmholtz Gemeinschaft

Search
Browse
Statistics
Feeds

(Phospho)proteomic profiling of microsatellite unstable CRC cells reveals alterations in nuclear signaling and cholesterol metabolism caused by frameshift mutation of NMD regulator UPF3A

[img]
Preview
PDF (Original Article) - Requires a PDF viewer such as GSview, Xpdf or Adobe Acrobat Reader
2MB
[img] Other (Supplementary Material)
2MB

Item Type:Article
Title:(Phospho)proteomic profiling of microsatellite unstable CRC cells reveals alterations in nuclear signaling and cholesterol metabolism caused by frameshift mutation of NMD regulator UPF3A
Creators Name:Michalak, M. and Katzenmaier, E.M. and Roeckel, N. and Woerner, S.M. and Fuchs, V. and Warnken, U. and Yuan, Y.P. and Bork, P. and Neu-Yilik, G. and Kulozik, A. and von Knebel Doeberitz, M. and Kloor, M. and Kopitz, J. and Gebert, J.
Abstract:DNA mismatch repair-deficient colorectal cancers (CRCs) accumulate numerous frameshift mutations at repetitive sequences recognized as microsatellite instability (MSI). When coding mononucleotide repeats (cMNRs) are affected, tumors accumulate frameshift mutations and premature termination codons (PTC) potentially leading to truncated proteins. Nonsense-mediated RNA decay (NMD) can degrade PTC-containing transcripts and protect from such faulty proteins. As it also regulates normal transcripts and cellular physiology, we tested whether NMD genes themselves are targets of MSI frameshift mutations. A high frequency of cMNR frameshift mutations in the UPF3A gene was found in MSI CRC cell lines (67.7%), MSI colorectal adenomas (55%) and carcinomas (63%). In normal colonic crypts, UPF3A expression was restricted to single chromogranin A-positive cells. SILAC-based proteomic analysis of KM12 CRC cells revealed UPF3A-dependent down-regulation of several enzymes involved in cholesterol biosynthesis. Furthermore, reconstituted UPF3A expression caused alterations of 85 phosphosites in 52 phosphoproteins. Most of them (38/52, 73%) reside in nuclear phosphoproteins involved in regulation of gene expression and RNA splicing. Since UPF3A mutations can modulate the (phospho)proteomic signature and expression of enzymes involved in cholesterol metabolism in CRC cells, UPF3A may influence other processes than NMD and loss of UPF3A expression might provide a growth advantage to MSI CRC cells.
Keywords:Nonsense-Mediated RNA Decay, Coding Mononucleotide Repeats, DNA Mismatch Repair, MSI Tumorigenesis, UPF3A
Source:International Journal of Molecular Sciences
ISSN:1422-0067
Publisher:MDPI
Volume:21
Number:15
Page Range:5234
Date:23 July 2020
Official Publication:https://doi.org/10.3390/ijms21155234
PubMed:View item in PubMed

Repository Staff Only: item control page

Downloads

Downloads per month over past year

Open Access
MDC Library