NAPRT Silencing in FH-Deficient Renal Cell Carcinoma Confers Therapeutic Vulnerabilities via NAD+ Depletion.

IF 4.1 2区 医学 Q2 CELL BIOLOGY
Katelyn J Noronha, Karlie N Lucas, Sateja Paradkar, Joseph Edmonds, Sam Friedman, Matthew A Murray, Samantha Liu, Dipti P Sajed, Chana Sachs, Josh Spurrier, Mitch Raponi, Jiayu Liang, Hao Zeng, Ranjini K Sundaram, Brian Shuch, Juan C Vasquez, Ranjit S Bindra
{"title":"NAPRT Silencing in FH-Deficient Renal Cell Carcinoma Confers Therapeutic Vulnerabilities via NAD+ Depletion.","authors":"Katelyn J Noronha, Karlie N Lucas, Sateja Paradkar, Joseph Edmonds, Sam Friedman, Matthew A Murray, Samantha Liu, Dipti P Sajed, Chana Sachs, Josh Spurrier, Mitch Raponi, Jiayu Liang, Hao Zeng, Ranjini K Sundaram, Brian Shuch, Juan C Vasquez, Ranjit S Bindra","doi":"10.1158/1541-7786.MCR-23-1003","DOIUrl":null,"url":null,"abstract":"<p><p>Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by loss of function mutations in fumarate hydratase (FH) and results in an aggressive subtype of renal cell carcinoma with limited treatment options. Loss of FH leads to accumulation of fumarate, an oncometabolite that disrupts multiple cellular processes and drives tumor progression. High levels of fumarate inhibit alpha ketoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) enzymes, and can lead to global DNA hypermethylation. Here, we report patterns of hypermethylation in FH-mutant cell lines and tumor samples are associated with the silencing of nicotinate phosphoribosyl transferase (NAPRT), a rate-limiting enzyme in the Preiss-Handler pathway of NAD+ biosynthesis, in a subset of HLRCC cases. NAPRT is hypermethylated at a CpG island in the promoter in cell line models and patient samples, resulting in loss of NAPRT expression. We find that FH-deficient RCC models with loss of NAPRT expression, as well as other oncometabolite-producing cancer models that silence NAPRT, are extremely sensitive to nicotinamide phosphoribosyl transferase inhibitors (NAMPTi). NAPRT silencing was also associated with synergistic tumor cell killing with PARP inhibitors and NAMPTis, which was associated with effects on PAR-mediated DNA repair. Overall, our findings indicate that NAPRT silencing can be targeted in oncometabolite-producing cancers and elucidates how oncometabolite-associated hypermethylation can impact diverse cellular processes and lead to therapeutically relevant vulnerabilities in cancer cells. Implications: NAPRT is a novel biomarker for targeting NAD+ metabolism in FH-deficient HLRCCs with NAMPTis alone and targeting DNA repair processes with the combination of NAMPTis and PARP inhibitors.</p>","PeriodicalId":19095,"journal":{"name":"Molecular Cancer Research","volume":null,"pages":null},"PeriodicalIF":4.1000,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11445649/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular Cancer Research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1158/1541-7786.MCR-23-1003","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by loss of function mutations in fumarate hydratase (FH) and results in an aggressive subtype of renal cell carcinoma with limited treatment options. Loss of FH leads to accumulation of fumarate, an oncometabolite that disrupts multiple cellular processes and drives tumor progression. High levels of fumarate inhibit alpha ketoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) enzymes, and can lead to global DNA hypermethylation. Here, we report patterns of hypermethylation in FH-mutant cell lines and tumor samples are associated with the silencing of nicotinate phosphoribosyl transferase (NAPRT), a rate-limiting enzyme in the Preiss-Handler pathway of NAD+ biosynthesis, in a subset of HLRCC cases. NAPRT is hypermethylated at a CpG island in the promoter in cell line models and patient samples, resulting in loss of NAPRT expression. We find that FH-deficient RCC models with loss of NAPRT expression, as well as other oncometabolite-producing cancer models that silence NAPRT, are extremely sensitive to nicotinamide phosphoribosyl transferase inhibitors (NAMPTi). NAPRT silencing was also associated with synergistic tumor cell killing with PARP inhibitors and NAMPTis, which was associated with effects on PAR-mediated DNA repair. Overall, our findings indicate that NAPRT silencing can be targeted in oncometabolite-producing cancers and elucidates how oncometabolite-associated hypermethylation can impact diverse cellular processes and lead to therapeutically relevant vulnerabilities in cancer cells. Implications: NAPRT is a novel biomarker for targeting NAD+ metabolism in FH-deficient HLRCCs with NAMPTis alone and targeting DNA repair processes with the combination of NAMPTis and PARP inhibitors.

FH缺陷肾细胞癌中的NAPRT沉默通过NAD+耗竭产生治疗脆弱性。
遗传性乳糜尿和肾细胞癌(HLRCC)是由富马酸氢化酶(FH)功能缺失突变引起的,是一种侵袭性肾细胞癌亚型,治疗方案有限。富马酸氢化酶的缺失会导致富马酸盐的积累,而富马酸盐是一种副代谢产物,会破坏多种细胞过程并推动肿瘤进展。高浓度的富马酸会抑制α-酮戊二酸依赖性二氧酶,包括十-十一转位(TET)酶,并可导致全局性DNA高甲基化。在这里,我们报告了 FH 突变细胞系和肿瘤样本中的高甲基化模式与烟酸磷酸核糖转移酶(NAPRT)的沉默有关,NAPRT 是 HLRCC 亚群病例中 NAD+ 生物合成的 Preiss-Handler 途径中的一个限速酶。在细胞系模型和患者样本中,NAPRT 在启动子的一个 CpG 岛处发生了高甲基化,导致 NAPRT 表达缺失。我们发现,NAPRT表达缺失的FH缺陷RCC模型以及其他沉默了NAPRT的产生肿瘤甲胎蛋白的癌症模型对烟酰胺磷酸核糖转移酶抑制剂(NAMPTis)极为敏感。NAPRT 沉默还与多(ADP)核糖聚合酶抑制剂(PARPis)和 NAMPTis 协同杀伤肿瘤细胞有关,这与 PAR 介导的 DNA 修复效应有关。总之,我们的研究结果表明,NAPRT-沉默可作为产生肿瘤代谢物的癌症的靶点,并阐明了肿瘤代谢物相关的高甲基化如何影响多种细胞过程,并导致癌细胞中与治疗相关的脆弱性。意义:NAPRT 是一种新的生物标志物,可用于单独使用 NAMPTis 靶向 FH 缺失的 HLRCC 中的 NAD+ 代谢,以及结合使用 NAMPTis 和 PARPis 靶向 DNA 修复过程。
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 求助全文
来源期刊
Molecular Cancer Research
Molecular Cancer Research 医学-细胞生物学
CiteScore
9.90
自引率
0.00%
发文量
280
审稿时长
4-8 weeks
期刊介绍: Molecular Cancer Research publishes articles describing novel basic cancer research discoveries of broad interest to the field. Studies must be of demonstrated significance, and the journal prioritizes analyses performed at the molecular and cellular level that reveal novel mechanistic insight into pathways and processes linked to cancer risk, development, and/or progression. Areas of emphasis include all cancer-associated pathways (including cell-cycle regulation; cell death; chromatin regulation; DNA damage and repair; gene and RNA regulation; genomics; oncogenes and tumor suppressors; signal transduction; and tumor microenvironment), in addition to studies describing new molecular mechanisms and interactions that support cancer phenotypes. For full consideration, primary research submissions must provide significant novel insight into existing pathway functions or address new hypotheses associated with cancer-relevant biologic questions.
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信