PMID- 23618389 OWN - NLM STAT- PubMed-not-MEDLINE DCOM- 20130517 LR - 20240321 IS - 1478-811X (Print) IS - 1478-811X (Electronic) IS - 1478-811X (Linking) VI - 11 IP - 1 DP - 2013 Apr 24 TI - The role of LPA and YAP signaling in long-term migration of human ovarian cancer cells. PG - 31 LID - 10.1186/1478-811X-11-31 [doi] AB - BACKGROUND: The Hippo-YAP signaling pathway is altered and implicated as oncogenic in many human cancers. However, extracellular signals that regulate the mammalian Hippo pathway have remained elusive until very recently when it was shown that the Hippo pathway is regulated by G-protein-coupled receptor (GPCR) ligands including lysophosphatidic acid (LPA) and sphingosine 1-phosphophate (S1P). LPA inhibits Lats kinase activity in HEK293 cells, but the potential involvement of a protein phosphatase was not investigated. The extracellular regulators of YAP dephosphorylation (dpYAP) and nuclear translocation in epithelial ovarian cancer (EOC) are essentially unknown. RESULTS: We showed here that LPA dose- and time-dependently induced dpYAP in human EOC cell lines OVCA433, OVCAR5, CAOV3, and Monty-1, accompanied by increased YAP nuclear translocation. YAP was involved in LPA-induced migration and invasion of EOC cells and LPA3 was a major LPA receptor mediating the migratory effect. We demonstrated that G13, but not or to a lesser extent G12, Gi or Gq, was necessary for LPA-induced dpYAP and its nuclear translocation and that RhoA-ROCK, but not RhoB, RhoC, Rac1, cdc42, PI3K, ERK, or AKT, were required for the LPA-dpYAP effect. In contrast to results in HEK293 cells, LPA did not inhibit Mst and Lats kinase in OVCA433 EOC cells. Instead, protein phosphatase 1A (PP1A) acted down-stream of RhoA in LPA-induction of dpYAP. In addition, we identified that amphiregulin (AREG), a down-stream target of YAP which activated EGF receptors (EGFR), mediated an LPA-stimulated and EGFR-dependent long-term (16 hr) cell migration. This process was transcription- and translation-dependent and was distinct from a transcription- and YAP-independent short-term (4 hr) cell migration. EOC tissues had reduced pYAP levels compared to normal and benign ovarian tissues, implying the involvement of dpYAP in EOC pathogenesis, as well as its potential marker and/or target values. CONCLUSIONS: A novel LPA-LPA3-G13-RhoA-ROCK-PP1A-dpYAP-AREG-EGFR signaling pathway was linked to LPA-induced migration of EOC cells. Reduced pYAP levels were demonstrated in human EOC tumors as compared to both normal ovarian tissues and benign gynecologic masses. Our findings support that YAP is a potential marker and target for developing novel therapeutic strategies against EOC. FAU - Cai, Hui AU - Cai H AD - Department of Obstetrics and Gynecology, Indiana University School of Medicine, 975 W, Walnut St, IB355A, Indianapolis, IN 46202, USA. xu2@iupui.edu. FAU - Xu, Yan AU - Xu Y LA - eng PT - Journal Article DEP - 20130424 PL - England TA - Cell Commun Signal JT - Cell communication and signaling : CCS JID - 101170464 EIN - Cell Commun Signal. 2013;11:92 PMC - PMC3655373 EDAT- 2013/04/27 06:00 MHDA- 2013/04/27 06:01 PMCR- 2013/04/24 CRDT- 2013/04/27 06:00 PHST- 2013/01/10 00:00 [received] PHST- 2013/04/09 00:00 [accepted] PHST- 2013/04/27 06:00 [entrez] PHST- 2013/04/27 06:00 [pubmed] PHST- 2013/04/27 06:01 [medline] PHST- 2013/04/24 00:00 [pmc-release] AID - 1478-811X-11-31 [pii] AID - 10.1186/1478-811X-11-31 [doi] PST - epublish SO - Cell Commun Signal. 2013 Apr 24;11(1):31. doi: 10.1186/1478-811X-11-31.