1. Academic Validation
  2. Kindlin-2 promotes Src-mediated tyrosine phosphorylation of androgen receptor and contributes to breast cancer progression

Kindlin-2 promotes Src-mediated tyrosine phosphorylation of androgen receptor and contributes to breast cancer progression

  • Cell Death Dis. 2022 May 20;13(5):482. doi: 10.1038/s41419-022-04945-z.
Luyao Ma  # 1 Yeteng Tian  # 1 Tao Qian 1 Wenjun Li 1 Chengmin Liu 1 Bizhu Chu 2 Qian Kong 2 Renwei Cai 1 Panzhu Bai 1 Lisha Ma 1 Yi Deng 1 Ruijun Tian 2 Chuanyue Wu 3 Ying Sun 4
Affiliations

Affiliations

  • 1 Department of Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China.
  • 2 Department of Chemistry, Southern University of Science and Technology, Shenzhen, 518055, China.
  • 3 Department of Pathology, School of Medicine and University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15260, USA. carywu@pitt.edu.
  • 4 Department of Biology, School of Life Sciences, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, Shenzhen, 518055, China. suny@sustech.edu.cn.
  • # Contributed equally.
Abstract

Androgen Receptor (AR) signaling plays important roles in breast Cancer progression. We show here that Kindlin-2, a focal adhesion protein, is critically involved in the promotion of AR signaling and breast Cancer progression. Kindlin-2 physically associates with AR and Src through its two neighboring domains, namely F1 and F0 domains, resulting in formation of a Kindlin-2-AR-Src supramolecular complex and consequently facilitating Src-mediated AR Tyr-534 phosphorylation and signaling. Depletion of Kindlin-2 was sufficient to suppress Src-mediated AR Tyr-534 phosphorylation and signaling, resulting in diminished breast Cancer cell proliferation and migration. Re-expression of wild-type Kindlin-2, but not AR-binding-defective or Src-binding-defective mutant forms of Kindlin-2, in Kindlin-2-deficient cells restored AR Tyr-534 phosphorylation, signaling, breast Cancer cell proliferation and migration. Furthermore, re-introduction of phosphor-mimic mutant AR-Y534D, but not wild-type AR reversed Kindlin-2 deficiency-induced inhibition of AR signaling and breast Cancer progression. Finally, using a genetic knockout strategy, we show that ablation of Kindlin-2 from mammary tumors in mouse significantly reduced AR Tyr-534 phosphorylation, breast tumor progression and metastasis in vivo. Our results suggest a critical role of Kindlin-2 in promoting breast Cancer progression and shed LIGHT on the molecular mechanism through which it functions in this process.

Figures
Products