β-Catenin/Tcf-4 inhibition after progastrin targeting reduces growth and drives differentiation of intestinal tumors

J Pannequin, N Delaunay, M Buchert, F Surrel… - Gastroenterology, 2007 - Elsevier
J Pannequin, N Delaunay, M Buchert, F Surrel, JF Bourgaux, J Ryan, S Boireau, J Coelho…
Gastroenterology, 2007Elsevier
Background & Aims: Aberrant activation of the β-catenin/Tcf-4 transcriptional complex
represents an initiating event for colorectal carcinogenesis, shifting the balance from
differentiation toward proliferation in colonic crypts. Here, we assessed whether
endogenous progastrin, encoded by a target gene of this complex, was in turn able to
regulate β-catenin/Tcf-4 activity in adenomatous polyposis coli (APC)-mutated cells, and we
analyzed the impact of topical progastrin depletion on intestinal tumor growth in vivo …
Background & Aims
Aberrant activation of the β-catenin/Tcf-4 transcriptional complex represents an initiating event for colorectal carcinogenesis, shifting the balance from differentiation toward proliferation in colonic crypts. Here, we assessed whether endogenous progastrin, encoded by a target gene of this complex, was in turn able to regulate β-catenin/Tcf-4 activity in adenomatous polyposis coli (APC)-mutated cells, and we analyzed the impact of topical progastrin depletion on intestinal tumor growth in vivo.
Methods
Stable or transient RNA silencing of the GAST gene was induced in human tumor cells and in mice carrying a heterozygous Apc mutation (APCΔ14), which overexpress progastrin but not amidated or glycine-extended gastrin.
Results
Depletion of endogenous progastrin production strongly decreased intestinal tumor growth in vivo through a marked inhibition of constitutive β-catenin/Tcf-4 activity in tumor cells. This effect was mediated by the de novo expression of the inhibitor of β-catenin and Tcf-4 (ICAT), resulting from a down-regulation of integrin-linked kinase in progastrin-depleted cells. Accordingly, ICAT down-regulation was correlated with progastrin overexpression and Tcf-4 target gene activation in human colorectal tumors, and ICAT repression was detected in the colon epithelium of tumor-prone, progastrin-overexpressing mice. In APCΔ14 mice, small interfering RNA–mediated progastrin depletion not only reduced intestinal tumor size and numbers, but also increased goblet cell lineage differentiation and cell apoptosis in the remaining adenomas.
Conclusions
Thus, depletion of endogenous progastrin inhibits the tumorigenicity of APC-mutated colorectal cancer cells in vivo by promoting ICAT expression, thereby counteracting Tcf-4 activity. Progastrin targeting strategies should provide an exciting prospect for the differentiation therapy of colorectal cancer.
Elsevier