Sphingosine-1-Phosphate Inhibition Increases Endoplasmic Reticulum Stress to Enhance Oxaliplatin Sensitivity in Pancreatic Cancer.

IF 2.1 Q3 ONCOLOGY World Journal of Oncology Pub Date : 2024-04-01 Epub Date: 2024-03-21 DOI:10.14740/wjon1768
Zachary Gao, Harinarayanan Janakiraman, Yang Xiao, Sung Wook Kang, Jiangling Dong, Jasmine Choi, Besim Ogretmen, Hyun-Sung Lee, Ernest Ramsay Camp
{"title":"Sphingosine-1-Phosphate Inhibition Increases Endoplasmic Reticulum Stress to Enhance Oxaliplatin Sensitivity in Pancreatic Cancer.","authors":"Zachary Gao, Harinarayanan Janakiraman, Yang Xiao, Sung Wook Kang, Jiangling Dong, Jasmine Choi, Besim Ogretmen, Hyun-Sung Lee, Ernest Ramsay Camp","doi":"10.14740/wjon1768","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer resistant to current therapies, including oxaliplatin (Oxa). Growing evidence supports the ability of cancers to harness sphingolipid metabolism for survival. Sphingosine-1-phosphate (S1P) is an anti-apoptotic, pro-survival mediator that can influence cellular functions such as endoplasmic reticulum (ER) stress. We hypothesize that PDAC drives dysregulated sphingolipid metabolism and that S1P inhibition can enhance ER stress to improve therapeutic response to Oxa in PDAC.</p><p><strong>Methods: </strong>RNA sequencing data of sphingolipid mediators from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression Project (GTEx) datasets were analyzed. Murine and human PDAC cell lines were treated with small interfering RNA (siRNA) against sphingosine kinase-2 (SPHK2) or ABC294640 (ABC) and incubated with combinations of vehicle control or Oxa. In an orthotopic syngeneic KPC PDAC model, tumors were treated with either vehicle control, Oxa, ABC, or combination therapy.</p><p><strong>Results: </strong>RNA sequencing analysis revealed multiple significantly differentially expressed sphingolipid mediators (P < 0.05). <i>In vitro</i>, both siRNA knockdown of SPHK2 and ABC sensitized cells to Oxa therapy (P < 0.05), and induced eukaryotic initiation factor 2α (eIF2α) and protein kinase RNA-like endoplasmic reticulum kinase (PERK) phosphorylation, hallmarks of ER stress. <i>In vitro</i> therapy also increased extracellular high mobility group box 1 (HMGB1) release (P < 0.05), necessary for immunogenic cell death (ICD). <i>In vivo</i> combination therapy increased apoptotic markers as well as the intensity of HMGB1 staining compared to control (P < 0.05).</p><p><strong>Conclusions: </strong>Our evidence suggests that sphingolipid metabolism is dysregulated in PDAC. Furthermore, S1P inhibition can sensitize PDAC to Oxa therapy through increasing ER stress and can potentiate ICD induction. This highlights a potential therapeutic target for chemosensitizing PDAC as well as an adjunct for future chemoimmunotherapy strategies.</p>","PeriodicalId":46797,"journal":{"name":"World Journal of Oncology","volume":null,"pages":null},"PeriodicalIF":2.1000,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10965266/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"World Journal of Oncology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.14740/wjon1768","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/3/21 0:00:00","PubModel":"Epub","JCR":"Q3","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer resistant to current therapies, including oxaliplatin (Oxa). Growing evidence supports the ability of cancers to harness sphingolipid metabolism for survival. Sphingosine-1-phosphate (S1P) is an anti-apoptotic, pro-survival mediator that can influence cellular functions such as endoplasmic reticulum (ER) stress. We hypothesize that PDAC drives dysregulated sphingolipid metabolism and that S1P inhibition can enhance ER stress to improve therapeutic response to Oxa in PDAC.

Methods: RNA sequencing data of sphingolipid mediators from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression Project (GTEx) datasets were analyzed. Murine and human PDAC cell lines were treated with small interfering RNA (siRNA) against sphingosine kinase-2 (SPHK2) or ABC294640 (ABC) and incubated with combinations of vehicle control or Oxa. In an orthotopic syngeneic KPC PDAC model, tumors were treated with either vehicle control, Oxa, ABC, or combination therapy.

Results: RNA sequencing analysis revealed multiple significantly differentially expressed sphingolipid mediators (P < 0.05). In vitro, both siRNA knockdown of SPHK2 and ABC sensitized cells to Oxa therapy (P < 0.05), and induced eukaryotic initiation factor 2α (eIF2α) and protein kinase RNA-like endoplasmic reticulum kinase (PERK) phosphorylation, hallmarks of ER stress. In vitro therapy also increased extracellular high mobility group box 1 (HMGB1) release (P < 0.05), necessary for immunogenic cell death (ICD). In vivo combination therapy increased apoptotic markers as well as the intensity of HMGB1 staining compared to control (P < 0.05).

Conclusions: Our evidence suggests that sphingolipid metabolism is dysregulated in PDAC. Furthermore, S1P inhibition can sensitize PDAC to Oxa therapy through increasing ER stress and can potentiate ICD induction. This highlights a potential therapeutic target for chemosensitizing PDAC as well as an adjunct for future chemoimmunotherapy strategies.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
抑制鞘氨醇-1-磷酸可增加内质网应激,提高胰腺癌对奥沙利铂的敏感性
背景:胰腺导管腺癌(PDAC胰腺导管腺癌(PDAC)是一种侵袭性癌症,对包括奥沙利铂(Oxa)在内的现有疗法具有抗药性。越来越多的证据表明,癌症有能力利用鞘脂代谢来维持生存。鞘磷脂-1-磷酸(S1P)是一种抗凋亡、促生存的介质,可影响细胞功能,如内质网(ER)应激。我们假设 PDAC 驱动鞘脂代谢失调,而抑制 S1P 可增强 ER 应激,从而改善 PDAC 对 Oxa 的治疗反应:分析了癌症基因组图谱(TCGA)和基因型-组织表达项目(GTEx)数据集中鞘脂介质的RNA测序数据。用针对鞘磷脂激酶-2(SPHK2)或 ABC294640(ABC)的小干扰 RNA(siRNA)处理小鼠和人类 PDAC 细胞系,并与载体对照或 Oxa 结合培养。在正位合成 KPC PDAC 模型中,肿瘤接受药物对照、Oxa、ABC 或联合疗法的治疗:结果:RNA测序分析表明,多种鞘脂介质的表达存在显著差异(P < 0.05)。在体外,siRNA 敲除 SPHK2 和 ABC 可使细胞对 Oxa 治疗敏感(P < 0.05),并诱导真核启动因子 2α (eIF2α)和蛋白激酶 RNA 样内质网激酶(PERK)磷酸化,这是 ER 应激的标志。体外疗法也增加了细胞外高迁移率基团框 1(HMGB1)的释放(P < 0.05),这是免疫原性细胞死亡(ICD)所必需的。与对照组相比,体内联合疗法增加了凋亡标志物以及 HMGB1 染色强度(P < 0.05):我们的证据表明,鞘脂代谢在PDAC中失调。此外,S1P抑制可通过增加ER应激使PDAC对Oxa治疗敏感,并可增强ICD诱导。这凸显了化疗增敏 PDAC 的潜在治疗靶点以及未来化疗免疫疗法策略的辅助手段。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
6.10
自引率
15.40%
发文量
37
期刊介绍: World Journal of Oncology, bimonthly, publishes original contributions describing basic research and clinical investigation of cancer, on the cellular, molecular, prevention, diagnosis, therapy and prognosis aspects. The submissions can be basic research or clinical investigation oriented. This journal welcomes those submissions focused on the clinical trials of new treatment modalities for cancer, and those submissions focused on molecular or cellular research of the oncology pathogenesis. Case reports submitted for consideration of publication should explore either a novel genomic event/description or a new safety signal from an oncolytic agent. The areas of interested manuscripts are these disciplines: tumor immunology and immunotherapy; cancer molecular pharmacology and chemotherapy; drug sensitivity and resistance; cancer epidemiology; clinical trials; cancer pathology; radiobiology and radiation oncology; solid tumor oncology; hematological malignancies; surgical oncology; pediatric oncology; molecular oncology and cancer genes; gene therapy; cancer endocrinology; cancer metastasis; prevention and diagnosis of cancer; other cancer related subjects. The types of manuscripts accepted are original article, review, editorial, short communication, case report, letter to the editor, book review.
期刊最新文献
Hereditary Gastric Cancer Is Linked With Hereditary Breast and Ovarian Cancer. High Probability of Lynch Syndrome Among Colorectal Cancer Patients Is Associated With Higher Occurrence of KRAS and PIK3CA Mutations. Identification and Validation of a Novel Tertiary Lymphoid Structures-Related Prognostic Gene Signature in Hepatocellular Carcinoma. Impact of Lymph Node Dissection for Patients With Clinically Node-Negative Intrahepatic Cholangiocarcinoma: A Multicenter Cohort Study. Increased Vitamin C Intake Is Associated With Decreased Pancreatic Cancer Risk.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1