Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer Cells

K-ras (Kirsten ras GTPase) mutations are oncogenic events frequently observed in many cancer types especially in pancreatic cancer. Although mitochondrial dysfunction has been associated with K-ras mutation, the molecular mechanisms by which K-ras impacts mitochondria and maintains metabolic homeost...

Full description

Bibliographic Details
Main Authors: Ning Meng, Christophe Glorieux, Yanyu Zhang, Liyun Liang, Peiting Zeng, Wenhua Lu, Peng Huang
Format: Article
Language:English
Published: MDPI AG 2019-12-01
Series:Cancers
Subjects:
Online Access:https://www.mdpi.com/2072-6694/12/1/65
_version_ 1797724635752562688
author Ning Meng
Christophe Glorieux
Yanyu Zhang
Liyun Liang
Peiting Zeng
Wenhua Lu
Peng Huang
author_facet Ning Meng
Christophe Glorieux
Yanyu Zhang
Liyun Liang
Peiting Zeng
Wenhua Lu
Peng Huang
author_sort Ning Meng
collection DOAJ
description K-ras (Kirsten ras GTPase) mutations are oncogenic events frequently observed in many cancer types especially in pancreatic cancer. Although mitochondrial dysfunction has been associated with K-ras mutation, the molecular mechanisms by which K-ras impacts mitochondria and maintains metabolic homeostasis are not fully understood. In this study, we used two K-ras inducible cell systems, human pancreatic epithelial/ K-ras<sup>G12D</sup> (HPNE/K-ras<sup>G12D</sup>) and human embryonic kidney cells with tetracycline repressorT-Rex/K-ras<sup>G12V</sup>, to evaluate the role of oncogenic K-ras in regulating mitochondrial function. Among a panel of genes known to affect mitochondria, only the expression of OPA3 (optic atrophy protein 3) was consistently up-regulated by K-ras activation in both cell lines. Importantly, high expression of OPA3 was also observed in clinical pancreatic cancer tissues. Genetic knockdown of OPA3 caused a significant decrease of energy metabolism, manifested by a suppression of oxygen consumption rate (OCR) and a decrease in cellular ATP content, leading to inhibition of cell proliferation capacity and reduced expression of epithelial&#8722;mesenchymal transition (EMT) markers. Our study suggests that OPA3 may promote cellular energy metabolism and its up-regulation in K-ras-driven cancer is likely a mechanism to offset the negative impact of K-ras on mitochondria to maintain energy homeostasis. As such, OPA3 could be a potential target to kill cancer cells with K-ras mutations.
first_indexed 2024-03-12T10:19:52Z
format Article
id doaj.art-a433f2f22cb5446d9eceb892fc6e4182
institution Directory Open Access Journal
issn 2072-6694
language English
last_indexed 2024-03-12T10:19:52Z
publishDate 2019-12-01
publisher MDPI AG
record_format Article
series Cancers
spelling doaj.art-a433f2f22cb5446d9eceb892fc6e41822023-09-02T10:11:08ZengMDPI AGCancers2072-66942019-12-011216510.3390/cancers12010065cancers12010065Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer CellsNing Meng0Christophe Glorieux1Yanyu Zhang2Liyun Liang3Peiting Zeng4Wenhua Lu5Peng Huang6Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, ChinaSun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, ChinaSun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, ChinaSun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, ChinaSun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, ChinaSun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, ChinaSun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, ChinaK-ras (Kirsten ras GTPase) mutations are oncogenic events frequently observed in many cancer types especially in pancreatic cancer. Although mitochondrial dysfunction has been associated with K-ras mutation, the molecular mechanisms by which K-ras impacts mitochondria and maintains metabolic homeostasis are not fully understood. In this study, we used two K-ras inducible cell systems, human pancreatic epithelial/ K-ras<sup>G12D</sup> (HPNE/K-ras<sup>G12D</sup>) and human embryonic kidney cells with tetracycline repressorT-Rex/K-ras<sup>G12V</sup>, to evaluate the role of oncogenic K-ras in regulating mitochondrial function. Among a panel of genes known to affect mitochondria, only the expression of OPA3 (optic atrophy protein 3) was consistently up-regulated by K-ras activation in both cell lines. Importantly, high expression of OPA3 was also observed in clinical pancreatic cancer tissues. Genetic knockdown of OPA3 caused a significant decrease of energy metabolism, manifested by a suppression of oxygen consumption rate (OCR) and a decrease in cellular ATP content, leading to inhibition of cell proliferation capacity and reduced expression of epithelial&#8722;mesenchymal transition (EMT) markers. Our study suggests that OPA3 may promote cellular energy metabolism and its up-regulation in K-ras-driven cancer is likely a mechanism to offset the negative impact of K-ras on mitochondria to maintain energy homeostasis. As such, OPA3 could be a potential target to kill cancer cells with K-ras mutations.https://www.mdpi.com/2072-6694/12/1/65k-rasmitochondriaopa3pancreatic cancerenergy metabolism
spellingShingle Ning Meng
Christophe Glorieux
Yanyu Zhang
Liyun Liang
Peiting Zeng
Wenhua Lu
Peng Huang
Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer Cells
Cancers
k-ras
mitochondria
opa3
pancreatic cancer
energy metabolism
title Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer Cells
title_full Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer Cells
title_fullStr Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer Cells
title_full_unstemmed Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer Cells
title_short Oncogenic K-ras Induces Mitochondrial OPA3 Expression to Promote Energy Metabolism in Pancreatic Cancer Cells
title_sort oncogenic k ras induces mitochondrial opa3 expression to promote energy metabolism in pancreatic cancer cells
topic k-ras
mitochondria
opa3
pancreatic cancer
energy metabolism
url https://www.mdpi.com/2072-6694/12/1/65
work_keys_str_mv AT ningmeng oncogenickrasinducesmitochondrialopa3expressiontopromoteenergymetabolisminpancreaticcancercells
AT christopheglorieux oncogenickrasinducesmitochondrialopa3expressiontopromoteenergymetabolisminpancreaticcancercells
AT yanyuzhang oncogenickrasinducesmitochondrialopa3expressiontopromoteenergymetabolisminpancreaticcancercells
AT liyunliang oncogenickrasinducesmitochondrialopa3expressiontopromoteenergymetabolisminpancreaticcancercells
AT peitingzeng oncogenickrasinducesmitochondrialopa3expressiontopromoteenergymetabolisminpancreaticcancercells
AT wenhualu oncogenickrasinducesmitochondrialopa3expressiontopromoteenergymetabolisminpancreaticcancercells
AT penghuang oncogenickrasinducesmitochondrialopa3expressiontopromoteenergymetabolisminpancreaticcancercells