Importantly, conditioned media from p16-defective cells stimulate

Importantly, conditioned media from p16-defective cells stimulated the invasion and the migration of cultured human epithelial cells. These results clearly show the role of the breast stromal fibroblast p16 protein in suppressing tumoregenesis. Moreover, we have shown that curcumin can normalize p16 expression and therefore reduces the expression and the secretion of these cancer promoting factors. This indicates that curcumin has potential ALK inhibitor use as stromal fibroblast normalizing factor

that can be utilized for the inhibition of both cancer initiation and recurrence. Hawsawi, N. M., Ghebeh, H., Hendrayani, S. F., Tulbah, A., Al-Eid, M., Al-Tweigeri, T., Ajarim, D., Alaiya, A., Dermime, S., and Aboussekhra, A. (2008). Cancer Res 68, 2717–2725. O95 Role of Heparanase in Colitis Associated Cancer Immanuel Lerner1, Eyal Zcharia1, Esther Bensoussan1, Dina Rodkin1, Yoav Sherman2, Israel Vlodavsky3, Michael Elkin 1 1 Department

of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel, 2 Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel, 3 Cancer and Vascular Biology Center, The Rappaport Selumetinib Faculty of Medicine, Haifa, Israel Ulcerative colitis (UC) is a chronic inflammatory bowel disease that is closely associated with colon cancer. Here we report that heparanase enzyme acts as an important mediator of colitis-associated tumorigenesis. Heparanase is an only known mammalian enzyme that cleaves heparan sulfate, the major polysaccharide of the extracellular matrix, and plays multiple roles in inflammation

and cancer progression. Applying histological specimens from UC patients and a mouse model of dextran sulfate sodium (DSS)-induced Enzalutamide colitis, we found that heparanase is constantly overexpressed and activated during the course of the disease, both in the active and inactive phases of inflammation. Employing heparanase-overexpressing transgenic mice in the model of colitis-associated cancer, induced by carcinogen azoxymethane followed by repeated DSS administration, we demonstrated that heparanase overexpression markedly increased the incidence and severity of colitis-associated colonic tumors, enabling faster tumor take, angiogenic switch and enhanced tumor progression. Notably, DSS-induced colitis alone (without azoxymethane pretreatment) lead to formation of colonic tumors in heparanase-transgenic, but not wild type mice, positioning heparanase as important physiological determinant in inflammation-driven colon carcinoma, replacing the need for carcinogen. Investigating molecular mechanisms underlying heparanase induction in colitis, we found that TNFalfa is responsible for continuous overexpression of heparanase by chronically-inflamed colonic epithelium.

Comments are closed.