Abstract

Abstract Introduction: Gastric cancer is the second most common cause of cancer-related death worldwide. Altered metabolism is considered a primary hallmark of tumorigenesis, as it can regulate important processes associated with proliferation, migration, and invasion. Lysophosphatidic acid (LPA), a multifunctional endogenous phospholipid, plays a vital role in cellular homeostasis and malignant behavior of the cancer cells through G-protein coupled receptors. Although several signaling pathways have been reported as a critical mediator of gastric cancer, LPA-LPAR2 axis-mediated alteration of energy metabolism in gastric cancer progression has not been established yet. Therefore, our study focused on the molecular mechanism of LPA-LPAR2 axis-mediated alteration of mitochondrial bioenergetics during gastric cancer progression. Method: TCGA database analysis was carried out to check the mRNA levels of the LPA receptors. Expression of LPA receptors was investigated in human gastric cancer samples by immunoblotting, RT-PCR, and immunohistochemical analysis. We elucidated the functional effects of LPA on gastric cancer by performing ECIS proliferation, migration assay, scratch assay, and transwell invasion assay in gastric cancer cells. To confirm the involvement of the specific LPA receptor on gastric cancer progression, we treated the cells with LPA1-3 receptor blocker (Ki16425) and LPA2 receptor-specific antagonist. Following LPA receptor blockade, we analyzed the glycolytic and mitochondrial functions by measuring extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) using a Seahorse XF24 analyzer. Result: Our TCGA dataset analysis, LPA ELISA, and the Western blotting result showed the presence of LPA receptors in human gastric cancer tissues, specifically LPAR2 was robustly increased (P<0.001) in human gastric cancer tissue samples, and LPA level was significantly higher (P<0.001), suggesting a possible role of LPA in gastric cancer. LPA treatment increased proliferation, migration, and invasion activity in AGS and NCI-N87 gastric cancer cell lines. In contrast, LPA receptor antagonist Ki16425 abrogated the LPA-induced effect. Our seahorse analysis results showed that LPA treatment increased the OCR (P<0.001) and the ECAR (P<0.001), whereas LPAR2 specific receptor antagonists abrogated this effect (P<0.001). Conclusion: Together, our results suggested that the LPA-LPAR2 axis mediates gastric cancer initiation and progression by increasing energy metabolism via oxidative phosphorylation and glycolysis. Thus, targeting the LPAR2 receptor may give us a novel therapeutic approach to treat gastric cancer. Citation Format: Hosne Ara, Susmita Bhattarai, Sudha Sharma, Utsab Subedi, Xiuping Yu, Md. Shenuarin Bhuiyan, Sumitra Miriyala, Manikandan Panchatcharam. LPAR2 depletion suppresses gastric cancer via alteration of mitochondrial energy metabolism [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5810.

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