Enhanced sensitivity to IGF-II signaling links loss of imprinting of IGF2 to increased cell proliferation and tumor risk

A Kaneda, CJ Wang, R Cheong… - Proceedings of the …, 2007 - National Acad Sciences
A Kaneda, CJ Wang, R Cheong, W Timp, P Onyango, B Wen, CA Iacobuzio-Donahue
Proceedings of the National Academy of Sciences, 2007National Acad Sciences
Loss of imprinting (LOI) of the insulin-like growth factor-II gene (IGF2), leading to abnormal
activation of the normally silent maternal allele, is a common human epigenetic population
variant associated with a 5-fold increased frequency of colorectal neoplasia. Here, we show
first that LOI leads specifically to increased expression of proliferation-related genes in
mouse intestinal crypts. Surprisingly, LOI (+) mice also have enhanced sensitivity to IGF-II
signaling, not simply increased IGF-II levels, because in vivo blockade with NVP-AEW541, a …
Loss of imprinting (LOI) of the insulin-like growth factor-II gene (IGF2), leading to abnormal activation of the normally silent maternal allele, is a common human epigenetic population variant associated with a 5-fold increased frequency of colorectal neoplasia. Here, we show first that LOI leads specifically to increased expression of proliferation-related genes in mouse intestinal crypts. Surprisingly, LOI(+) mice also have enhanced sensitivity to IGF-II signaling, not simply increased IGF-II levels, because in vivo blockade with NVP-AEW541, a specific inhibitor of the IGF-II signaling receptor, showed reduction of proliferation-related gene expression to levels half that seen in LOI(−) mice. Signal transduction assays in microfluidic chips confirmed this enhanced sensitivity with marked augmentation of Akt/PKB signaling in LOI(+) cells at low doses of IGF-II, which was reduced in the presence of the inhibitor to levels below those found in LOI(−) cells, and was associated with increased expression of the IGF1 and insulin receptor genes. We exploited this increased IGF-II sensitivity to develop an in vivo chemopreventive strategy using the azoxymethane (AOM) mutagenesis model. LOI(+) mice treated with AOM showed a 60% increase in premalignant aberrant crypt foci (ACF) formation over LOI(−) mice. In vivo IGF-II blockade with NVP-AEW541 abrogated this effect, reducing ACF to a level 30% lower even than found in exposed LOI(−) mice. Thus, LOI increases cancer risk in a counterintuitive way, by increasing the sensitivity of the IGF-II signaling pathway itself, providing a previously undescribed epigenetic chemoprevention strategy in which cells with LOI are “IGF-II addicted” and undergo reduced tumorigenesis in the colon upon IGF-II pathway blockade.
National Acad Sciences