CXCR4/YY1 inhibition impairs VEGF network and angiogenesis during malignancy

F de Nigris, V Crudele, A Giovane… - Proceedings of the …, 2010 - National Acad Sciences
F de Nigris, V Crudele, A Giovane, A Casamassimi, A Giordano, HJ Garban, F Cacciatore…
Proceedings of the National Academy of Sciences, 2010National Acad Sciences
Tumor growth requires neoangiogenesis. VEGF is the most potent proangiogenic factor.
Dysregulation of hypoxia-inducible factor (HIF) or cytokine stimuli such as those involving
the chemokine receptor 4/stromal-derived cell factor 1 (CXCR4/SDF-1) axis are the major
cause of ectopic overexpression of VEGF in tumors. Although the CXCR4/SDF-1 pathway is
well characterized, the transcription factors executing the effector function of this signaling
are poorly understood. The multifunctional Yin Yang 1 (YY1) protein is highly expressed in …
Tumor growth requires neoangiogenesis. VEGF is the most potent proangiogenic factor. Dysregulation of hypoxia-inducible factor (HIF) or cytokine stimuli such as those involving the chemokine receptor 4/stromal-derived cell factor 1 (CXCR4/SDF-1) axis are the major cause of ectopic overexpression of VEGF in tumors. Although the CXCR4/SDF-1 pathway is well characterized, the transcription factors executing the effector function of this signaling are poorly understood. The multifunctional Yin Yang 1 (YY1) protein is highly expressed in different types of cancers and may regulate some cancer-related genes. The network involving CXCR4/YY1 and neoangiogenesis could play a major role in cancer progression. In this study we have shown that YY1 forms an active complex with HIF-1α at VEGF gene promoters and increases VEGF transcription and expression observed by RT-PCR, ELISA, and Western blot using two different antibodies against VEGFB. Long-term treatment with T22 peptide (a CXCR4/SDF-1 inhibitor) and YY1 silencing can reduce in vivo systemic neoangiogenesis (P < 0.01 and P < 0.05 vs. control, respectively) during metastasis. Moreover, using an in vitro angiogenesis assay, we observed that YY1 silencing led to a 60% reduction in branches (P < 0.01) and tube length (P < 0.02) and a 75% reduction in tube area (P < 0.001) compared with control cells. A similar reduction was observed using T22 peptide. We demonstrated that T22 peptide determines YY1 cytoplasmic accumulation by reducing its phosphorylation via down-regulation of AKT, identifying a crosstalk mechanism involving CXCR4/YY1. Thus, YY1 may represent a crucial molecular target for antiangiogenic therapy during cancer progression.
National Acad Sciences