Abstract

BackgroundWe have previously reported that a new intronless gene for casein kinase 2α (CK2α), CSNK2A3, is expressed in human cells. The promoter of the well-known CK2α, CSNK2A1, displays characteristics of a housekeeping gene, whereas CSNK2A3 has a characteristic of a regulated promoter with two TATA boxes and a CAAT box. GPR68, a family of the G protein-coupled receptors, is also known as ovarian cancer G protein-coupled receptor 1 (OGR1). In the current study, we analyzed the roles of CK2α genes and neutral endopeptidase (NEP), a key enzyme that influences a variety of malignancies, in the OGR1-induced inhibition of A549 cell migration.MethodsWe analyzed the transcript expressions of both the CK2α genes (CSNK2A1 and CSNK2A3) and NEP upon OGR1 overexpression. Protein expression of CK2α and NEP were also analyzed. We further elucidated the functional roles of both CK2α and NEP in the OGR1-induced inhibition of A549 cell migration in vitro using a wound-healing assay. We also analyzed the molecular mechanisms involved in the OGR1-induced inhibition of lung cancer cell migration.ResultsThe findings of this study showed that OGR1 upregulated the expression of CSNK2A3 but not CSNK2A1 in the A549 cells. The findings further suggested OGR1 also upregulates the expression of NEP. The OGR1-induced inhibition of A549 cell migration was abrogated completely by inhibition of CK2α activity, whereas partial abrogation (~ 30%) was observed in the presence of NEP inhibition. The results also revealed that OGR1 regulates CSNK2A3 via activation of Rac1/cdc42 and MAPKs pathways. CK2 is ubiquitously expressed, and in contrast, is believed to be a constitutively active enzyme, and its regulation appears to be independent of known second messengers.ConclusionIn the current study, we report for the first time the OGR1-induced regulation of CSNK2A3, CK2αP, and NEP in A549 cancer cells. Our study also decoded the downstream cellular proteins of OGR1 as well as the molecular mechanism involved in OGR1-induced inhibition of A549 cell migration. The findings of this research suggest the potential therapeutic targets to inhibit lung cancer progression.

Highlights

  • GPR68 was first cloned from an ovarian cancer cell

  • ovarian cancer G protein-coupled receptor 1 (OGR1) regulates the expression of CSNK2A3 and neutral endopeptidase (NEP) but not CSNK2A1 To investigate the downstream molecules involved in the OGR1-induced inhibition of cancer cell migration, OGR1 was over-expressed transiently in A549 cells for 48 h, and transcript expression of CSNK2A1, CSNK2A3, and NEP were analyzed using semiqPCR

  • The results showed that OGR1 strongly upregulates transcript expression of CSNK2A3 (CK2α intronless gene, CK2αP) and NEP

Read more

Summary

Introduction

GPR68 was first cloned from an ovarian cancer cell. it is known as ovarian cancer G protein-coupled receptor 1 (OGR1) [1]. Studies have reported that OGR1 inhibits breast and ovarian cancer cells in vitro when it is reexpressed in cancer cells [8, 9]. Our previous study revealed that OGR1 has an inhibitory effect on prostate cancer tumorigenesis when expressed in host/stromal cells using the OGR1-knockout TRAMP mice prostate cancer model [10]. Another study by Horman SR, et al reported that murine colon tumor implants in OGR1 knockout mice displayed delayed tumor growth [12]. These previous reports indicate that in contrast to OGR1’s tumorsuppressing role in tumor cells, host cell OGR1 may be involved in and/or required for tumor growth. We analyzed the roles of CK2α genes and neutral endopeptidase (NEP), a key enzyme that influences a variety of malignancies, in the OGR1-induced inhibition of A549 cell migration

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call