1. Academic Validation
  2. LPA1-mediated inhibition of CXCR4 attenuates CXCL12-induced signaling and cell migration

LPA1-mediated inhibition of CXCR4 attenuates CXCL12-induced signaling and cell migration

  • Cell Commun Signal. 2023 Sep 25;21(1):257. doi: 10.1186/s12964-023-01261-7.
Jong Min Hong 1 Jin-Woo Lee 1 Dong-Seung Seen 2 Jae-Yeon Jeong 3 Won-Ki Huh 4 5
Affiliations

Affiliations

  • 1 School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
  • 2 GPCR Therapeutics Inc, Gwanak-Gu, Seoul, 08790, Republic of Korea.
  • 3 GPCR Therapeutics Inc, Gwanak-Gu, Seoul, 08790, Republic of Korea. [email protected].
  • 4 School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea. [email protected].
  • 5 Institute of Microbiology, Seoul National University, Seoul, 08826, Republic of Korea. [email protected].
Abstract

Background: G protein-coupled receptor heteromerization is believed to exert dynamic regulatory impact on signal transduction. CXC Chemokine Receptor 4 (CXCR4) and its ligand CXCL12, both of which are overexpressed in many cancers, play a pivotal role in metastasis. Likewise, lysophosphatidic acid receptor 1 (LPA1) is implicated in Cancer cell proliferation and migration. In our preliminary study, we identified LPA1 as a prospective CXCR4 interactor. In the present study, we investigated in detail the formation of the CXCR4-LPA1 heteromer and characterized the unique molecular features and function of this heteromer.

Methods: We employed bimolecular fluorescence complementation, bioluminescence resonance energy transfer, and proximity ligation assays to demonstrate heteromerization between CXCR4 and LPA1. To elucidate the distinctive molecular characteristics and functional implications of the CXCR4-LPA1 heteromer, we performed various assays, including cAMP, BRET for G protein activation, β-arrestin recruitment, ligand binding, and transwell migration assays.

Results: We observed that CXCR4 forms heteromers with LPA1 in recombinant HEK293A cells and the human breast Cancer cell line MDA-MB-231. Coexpression of LPA1 with CXCR4 reduced CXCL12-mediated cAMP inhibition, ERK activation, Gαi/o activation, and β-arrestin recruitment, while CXCL12 binding to CXCR4 remained unaffected. In contrast, CXCR4 had no impact on LPA1-mediated signaling. The addition of lysophosphatidic acid (LPA) further hindered CXCL12-induced Gαi/o recruitment to CXCR4. LPA or alkyl-OMPT inhibited CXCL12-induced migration in various Cancer cells that endogenously express both CXCR4 and LPA1. Conversely, CXCL12-induced calcium signaling and migration were increased in LPAR1 knockout cells, and LPA1-selective antagonists enhanced CXCL12-induced Gαi/o signaling and cell migration in the parental MDA-MB-231 cells but not in LPA1-deficient cells. Ultimately, complete inhibition of cell migration toward CXCL12 and alkyl-OMPT was only achieved in the presence of both CXCR4 and LPA1 antagonists.

Conclusions: The presence and impact of CXCR4-LPA1 heteromers on CXCL12-induced signaling and cell migration have been evidenced across various cell lines. This discovery provides crucial insights into a valuable regulatory mechanism of CXCR4 through heteromerization. Moreover, our findings propose a therapeutic potential in combined CXCR4 and LPA1 inhibitors for Cancer and inflammatory diseases associated with these receptors, simultaneously raising concerns about the use of LPA1 antagonists alone for such conditions. Video Abstract.

Keywords

Cancer; Chemokine receptor 4; Chemotaxis; G protein-coupled receptor; GPCR heteromer; GPCR signaling; Inflammatory disease; Lysophosphatidic acid receptor 1.

Figures
Products