%0 Journal Article %T Eya3 promotes breast tumor¨Cassociated immune suppression via threonine phosphatase¨Cmediated PD-L1 upregulation %A Debashis Ghosh %A Heide L. Ford %A Hengbo Zhou %A James C. Costello %A Jared Klarquist %A Jill E. Slansky %A Lingdi Zhang %A Melanie Y. Vincent %A Pratyaydipta Rudra %A Rani K. Powers %A Rebecca L. Vartuli %A Ross M. Kedl %A Rui Zhao %J The Journal of Clinical Investigation %D 2018 %R 10.1172/JCI96784 %X Eya proteins are critical developmental regulators that are highly expressed in embryogenesis but downregulated after development. Amplification and/or re-expression of Eyas occurs in many tumor types. In breast cancer, Eyas regulate tumor progression by acting as transcriptional cofactors and tyrosine phosphatases. Intriguingly, Eyas harbor a separate threonine (Thr) phosphatase activity, which was previously implicated in innate immunity. Here we describe what we believe to be a novel role for Eya3 in mediating triple-negative breast cancer¨Cassociated immune suppression. Eya3 loss decreases tumor growth in immune-competent mice and is associated with increased numbers of infiltrated CD8+ T cells, which, when depleted, reverse the effects of Eya3 knockdown. Mechanistically, Eya3 utilizes its Thr phosphatase activity to dephosphorylate Myc at pT58, resulting in a stabilized form. We show that Myc is required for Eya3-mediated increases in PD-L1, and that rescue of PD-L1 in Eya3-knockdown cells restores tumor progression. Finally, we demonstrate that Eya3 significantly correlates with PD-L1 in human breast tumors, and that tumors expressing high levels of Eya3 have a decreased CD8+ T cell signature. Our data uncover a role for Eya3 in mediating tumor-associated immune suppression, and suggest that its inhibition may enhance checkpoint therapies %U https://www.jci.org/articles/view/96784