首页 > 最新文献

NAR Cancer最新文献

英文 中文
Current proteomics methods applicable to dissecting the DNA damage response. 适用于剖析 DNA 损伤反应的当前蛋白质组学方法。
Pub Date : 2023-05-19 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad020
Monita Muralidharan, Nevan J Krogan, Mehdi Bouhaddou, Minkyu Kim

The DNA damage response (DDR) entails reorganization of proteins and protein complexes involved in DNA repair. The coordinated regulation of these proteomic changes maintains genome stability. Traditionally, regulators and mediators of DDR have been investigated individually. However, recent advances in mass spectrometry (MS)-based proteomics enable us to globally quantify changes in protein abundance, post-translational modifications (PTMs), protein localization, and protein-protein interactions (PPIs) in cells. Furthermore, structural proteomics approaches, such as crosslinking MS (XL-MS), hydrogen/deuterium exchange MS (H/DX-MS), Native MS (nMS), provide large structural information of proteins and protein complexes, complementary to the data collected from conventional methods, and promote integrated structural modeling. In this review, we will overview the current cutting-edge functional and structural proteomics techniques that are being actively utilized and developed to help interrogate proteomic changes that regulate the DDR.

DNA 损伤应答(DDR)需要重组参与 DNA 修复的蛋白质和蛋白质复合物。这些蛋白质组变化的协调调控可维持基因组的稳定性。传统上,DDR 的调节因子和介导因子都是单独研究的。然而,基于质谱(MS)的蛋白质组学的最新进展使我们能够全面量化细胞中蛋白质丰度、翻译后修饰(PTMs)、蛋白质定位和蛋白质-蛋白质相互作用(PPIs)的变化。此外,交联质谱(XL-MS)、氢/氘交换质谱(H/DX-MS)、原生质谱(nMS)等结构蛋白质组学方法提供了大量蛋白质和蛋白质复合物的结构信息,与传统方法收集的数据相辅相成,促进了综合结构建模的发展。在本综述中,我们将概述目前正在积极利用和开发的前沿功能和结构蛋白质组学技术,以帮助研究调控 DDR 的蛋白质组变化。
{"title":"Current proteomics methods applicable to dissecting the DNA damage response.","authors":"Monita Muralidharan, Nevan J Krogan, Mehdi Bouhaddou, Minkyu Kim","doi":"10.1093/narcan/zcad020","DOIUrl":"10.1093/narcan/zcad020","url":null,"abstract":"<p><p>The DNA damage response (DDR) entails reorganization of proteins and protein complexes involved in DNA repair. The coordinated regulation of these proteomic changes maintains genome stability. Traditionally, regulators and mediators of DDR have been investigated individually. However, recent advances in mass spectrometry (MS)-based proteomics enable us to globally quantify changes in protein abundance, post-translational modifications (PTMs), protein localization, and protein-protein interactions (PPIs) in cells. Furthermore, structural proteomics approaches, such as crosslinking MS (XL-MS), hydrogen/deuterium exchange MS (H/DX-MS), Native MS (nMS), provide large structural information of proteins and protein complexes, complementary to the data collected from conventional methods, and promote integrated structural modeling. In this review, we will overview the current cutting-edge functional and structural proteomics techniques that are being actively utilized and developed to help interrogate proteomic changes that regulate the DDR.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/93/59/zcad020.PMC10198729.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9901367","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GSK-3484862 targets DNMT1 for degradation in cells. GSK-3484862靶向DNMT1用于细胞中的降解。
Pub Date : 2023-05-17 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad022
Qin Chen, Bigang Liu, Yang Zeng, Jee Won Hwang, Nan Dai, Ivan R Corrêa, Marcos R Estecio, Xing Zhang, Margarida A Santos, Taiping Chen, Xiaodong Cheng

Maintenance of genomic methylation patterns at DNA replication forks by DNMT1 is the key to faithful mitotic inheritance. DNMT1 is often overexpressed in cancer cells and the DNA hypomethylating agents azacytidine and decitabine are currently used in the treatment of hematologic malignancies. However, the toxicity of these cytidine analogs and their ineffectiveness in treating solid tumors have limited wider clinical use. GSK-3484862 is a newly-developed, dicyanopyridine containing, non-nucleoside DNMT1-selective inhibitor with low cellular toxicity. Here, we show that GSK-3484862 targets DNMT1 for protein degradation in both cancer cell lines and murine embryonic stem cells (mESCs). DNMT1 depletion was rapid, taking effect within hours following GSK-3484862 treatment, leading to global hypomethylation. Inhibitor-induced DNMT1 degradation was proteasome-dependent, with no discernible loss of DNMT1 mRNA. In mESCs, GSK-3484862-induced Dnmt1 degradation requires the Dnmt1 accessory factor Uhrf1 and its E3 ubiquitin ligase activity. We also show that Dnmt1 depletion and DNA hypomethylation induced by the compound are reversible after its removal. Together, these results indicate that this DNMT1-selective degrader/inhibitor will be a valuable tool for dissecting coordinated events linking DNA methylation to gene expression and identifying downstream effectors that ultimately regulate cellular response to altered DNA methylation patterns in a tissue/cell-specific manner.

DNMT1在DNA复制叉维持基因组甲基化模式是忠实有丝分裂遗传的关键。DNMT1通常在癌症细胞中过表达,DNA低甲基化剂氮胞苷和地西他滨目前用于治疗血液系统恶性肿瘤。然而,这些胞苷类似物的毒性及其在治疗实体瘤方面的无效性限制了其更广泛的临床应用。GSK-3484862是一种新开发的含双氰基吡啶的非核苷DNMT1选择性抑制剂,具有低细胞毒性。在此,我们发现GSK-3484862靶向DNMT1,用于癌症细胞系和小鼠胚胎干细胞(mESCs)中的蛋白质降解。DNMT1消耗迅速,在GSK-3484862治疗后数小时内生效,导致整体低甲基化。抑制剂诱导的DNMT1降解是蛋白酶体依赖性的,没有明显的DNMT1mRNA损失。在mESCs中,GSK-3484862诱导的Dnmt1降解需要Dnmt1辅助因子Uhrf1及其E3泛素连接酶活性。我们还表明,该化合物诱导的Dnmt1缺失和DNA低甲基化在其去除后是可逆的。总之,这些结果表明,这种DNMT1选择性降解物/抑制剂将是一种有价值的工具,用于分析将DNA甲基化与基因表达联系起来的协同事件,并鉴定最终以组织/细胞特异性方式调节细胞对改变的DNA甲基化模式的反应的下游效应物。
{"title":"GSK-3484862 targets DNMT1 for degradation in cells.","authors":"Qin Chen,&nbsp;Bigang Liu,&nbsp;Yang Zeng,&nbsp;Jee Won Hwang,&nbsp;Nan Dai,&nbsp;Ivan R Corrêa,&nbsp;Marcos R Estecio,&nbsp;Xing Zhang,&nbsp;Margarida A Santos,&nbsp;Taiping Chen,&nbsp;Xiaodong Cheng","doi":"10.1093/narcan/zcad022","DOIUrl":"10.1093/narcan/zcad022","url":null,"abstract":"<p><p>Maintenance of genomic methylation patterns at DNA replication forks by DNMT1 is the key to faithful mitotic inheritance. DNMT1 is often overexpressed in cancer cells and the DNA hypomethylating agents azacytidine and decitabine are currently used in the treatment of hematologic malignancies. However, the toxicity of these cytidine analogs and their ineffectiveness in treating solid tumors have limited wider clinical use. GSK-3484862 is a newly-developed, dicyanopyridine containing, non-nucleoside DNMT1-selective inhibitor with low cellular toxicity. Here, we show that GSK-3484862 targets DNMT1 for protein degradation in both cancer cell lines and murine embryonic stem cells (mESCs). DNMT1 depletion was rapid, taking effect within hours following GSK-3484862 treatment, leading to global hypomethylation. Inhibitor-induced DNMT1 degradation was proteasome-dependent, with no discernible loss of <i>DNMT1</i> mRNA. In mESCs, GSK-3484862-induced Dnmt1 degradation requires the Dnmt1 accessory factor Uhrf1 and its E3 ubiquitin ligase activity. We also show that Dnmt1 depletion and DNA hypomethylation induced by the compound are reversible after its removal. Together, these results indicate that this DNMT1-selective degrader/inhibitor will be a valuable tool for dissecting coordinated events linking DNA methylation to gene expression and identifying downstream effectors that ultimately regulate cellular response to altered DNA methylation patterns in a tissue/cell-specific manner.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-05-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/bd/6b/zcad022.PMC10189803.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9864768","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular mechanisms protecting centromeres from self-sabotage and implications for cancer therapy. 保护着丝粒免受自我破坏的分子机制及其对癌症治疗的意义。
Pub Date : 2023-05-09 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad019
Rim Nassar, Lily Thompson, Elise Fouquerel

Centromeres play a crucial role in DNA segregation by mediating the cohesion and separation of sister chromatids during cell division. Centromere dysfunction, breakage or compromised centromeric integrity can generate aneuploidies and chromosomal instability, which are cellular features associated with cancer initiation and progression. Maintaining centromere integrity is thus essential for genome stability. However, the centromere itself is prone to DNA breaks, likely due to its intrinsically fragile nature. Centromeres are complex genomic loci that are composed of highly repetitive DNA sequences and secondary structures and require the recruitment and homeostasis of a centromere-associated protein network. The molecular mechanisms engaged to preserve centromere inherent structure and respond to centromeric damage are not fully understood and remain a subject of ongoing research. In this article, we provide a review of the currently known factors that contribute to centromeric dysfunction and the molecular mechanisms that mitigate the impact of centromere damage on genome stability. Finally, we discuss the potential therapeutic strategies that could arise from a deeper understanding of the mechanisms preserving centromere integrity.

在细胞分裂过程中,着丝粒通过介导姐妹染色单体的内聚和分离,在DNA分离中发挥着至关重要的作用。着丝粒功能障碍、断裂或着丝粒完整性受损可产生非整倍体和染色体不稳定性,这是与癌症发生和发展相关的细胞特征。因此,保持着丝粒的完整性对基因组的稳定性至关重要。然而,着丝粒本身容易发生DNA断裂,这可能是由于其固有的脆弱性。着丝粒是由高度重复的DNA序列和二级结构组成的复杂基因组基因座,需要着丝粒相关蛋白网络的募集和稳态。保护着丝粒固有结构和对着丝粒损伤作出反应的分子机制尚不完全清楚,目前仍在进行研究。在这篇文章中,我们对目前已知的导致着丝粒功能障碍的因素以及减轻着丝粒损伤对基因组稳定性影响的分子机制进行了综述。最后,我们讨论了潜在的治疗策略,这些策略可能源于对保持着丝粒完整性的机制的更深入理解。
{"title":"Molecular mechanisms protecting centromeres from self-sabotage and implications for cancer therapy.","authors":"Rim Nassar,&nbsp;Lily Thompson,&nbsp;Elise Fouquerel","doi":"10.1093/narcan/zcad019","DOIUrl":"10.1093/narcan/zcad019","url":null,"abstract":"<p><p>Centromeres play a crucial role in DNA segregation by mediating the cohesion and separation of sister chromatids during cell division. Centromere dysfunction, breakage or compromised centromeric integrity can generate aneuploidies and chromosomal instability, which are cellular features associated with cancer initiation and progression. Maintaining centromere integrity is thus essential for genome stability. However, the centromere itself is prone to DNA breaks, likely due to its intrinsically fragile nature. Centromeres are complex genomic loci that are composed of highly repetitive DNA sequences and secondary structures and require the recruitment and homeostasis of a centromere-associated protein network. The molecular mechanisms engaged to preserve centromere inherent structure and respond to centromeric damage are not fully understood and remain a subject of ongoing research. In this article, we provide a review of the currently known factors that contribute to centromeric dysfunction and the molecular mechanisms that mitigate the impact of centromere damage on genome stability. Finally, we discuss the potential therapeutic strategies that could arise from a deeper understanding of the mechanisms preserving centromere integrity.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/20/a5/zcad019.PMC10167631.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9509927","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Therapeutic disruption of RAD52-ssDNA complexation via novel drug-like inhibitors. 通过新型药物抑制剂对 RAD52-ssDNA 复合物进行治疗性破坏。
Pub Date : 2023-05-01 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad018
Divya S Bhat, Eva Malacaria, Ludovica Di Biagi, Mortezaali Razzaghi, Masayoshi Honda, Kathryn F Hobbs, Sarah R Hengel, Pietro Pichierri, M Ashley Spies, Maria Spies

RAD52 protein is a coveted target for anticancer drug discovery. Similar to poly-ADP-ribose polymerase (PARP) inhibitors, pharmacological inhibition of RAD52 is synthetically lethal with defects in genome caretakers BRCA1 and BRCA2 (∼25% of breast and ovarian cancers). Emerging structure activity relationships for RAD52 are complex, making it challenging to transform previously identified disruptors of the RAD52-ssDNA interaction into drug-like leads using traditional medicinal chemistry approaches. Using pharmacophoric informatics on the RAD52 complexation by epigallocatechin (EGC), and the Enamine in silico REAL database, we identified six distinct chemical scaffolds that occupy the same physical space on RAD52 as EGC. All six were RAD52 inhibitors (IC50 ∼23-1200 μM) with two of the compounds (Z56 and Z99) selectively killing BRCA-mutant cells and inhibiting cellular activities of RAD52 at micromolar inhibitor concentrations. While Z56 had no effect on the ssDNA-binding protein RPA and was toxic to BRCA-mutant cells only, Z99 inhibited both proteins and displayed toxicity towards BRCA-complemented cells. Optimization of the Z99 scaffold resulted in a set of more powerful and selective inhibitors (IC50 ∼1.3-8 μM), which were only toxic to BRCA-mutant cells. RAD52 complexation by Z56, Z99 and its more specific derivatives provide a roadmap for next generation of cancer therapeutics.

RAD52 蛋白是人们梦寐以求的抗癌药物靶点。与多聚 ADP 核糖聚合酶(PARP)抑制剂类似,RAD52 的药理抑制也会导致基因组看护者 BRCA1 和 BRCA2 的缺陷(占乳腺癌和卵巢癌的 25%)。新发现的 RAD52 结构活性关系非常复杂,因此使用传统的药物化学方法将以前发现的 RAD52 与 ssDNA 相互作用的干扰物转化为类似药物的先导物具有挑战性。利用表没食子儿茶素(EGC)与 RAD52 复合物的药理学信息学以及 Enamine in silico REAL 数据库,我们发现了与 EGC 在 RAD52 上占据相同物理空间的六种不同的化学支架。这六种化合物都是 RAD52 抑制剂(IC50 ∼23-1200 μM),其中两种化合物(Z56 和 Z99)能选择性地杀死 BRCA 突变细胞,并在微摩尔抑制剂浓度下抑制 RAD52 的细胞活性。Z56对ssDNA结合蛋白RPA没有影响,仅对BRCA突变细胞有毒性,而Z99对两种蛋白都有抑制作用,并对BRCA补体细胞有毒性。对 Z99 支架的优化产生了一组更强、更有选择性的抑制剂(IC50 ∼1.3-8 μM),它们只对 BRCA 突变细胞有毒性。Z56、Z99 及其更具特异性的衍生物对 RAD52 的复合物作用为下一代癌症疗法提供了路线图。
{"title":"Therapeutic disruption of RAD52-ssDNA complexation via novel drug-like inhibitors.","authors":"Divya S Bhat, Eva Malacaria, Ludovica Di Biagi, Mortezaali Razzaghi, Masayoshi Honda, Kathryn F Hobbs, Sarah R Hengel, Pietro Pichierri, M Ashley Spies, Maria Spies","doi":"10.1093/narcan/zcad018","DOIUrl":"10.1093/narcan/zcad018","url":null,"abstract":"<p><p>RAD52 protein is a coveted target for anticancer drug discovery. Similar to poly-ADP-ribose polymerase (PARP) inhibitors, pharmacological inhibition of RAD52 is synthetically lethal with defects in genome caretakers BRCA1 and BRCA2 (∼25% of breast and ovarian cancers). Emerging structure activity relationships for RAD52 are complex, making it challenging to transform previously identified disruptors of the RAD52-ssDNA interaction into drug-like leads using traditional medicinal chemistry approaches. Using pharmacophoric informatics on the RAD52 complexation by epigallocatechin (EGC), and the Enamine <i>in silico</i> REAL database, we identified six distinct chemical scaffolds that occupy the same physical space on RAD52 as EGC. All six were RAD52 inhibitors (IC<sub>50</sub> ∼23-1200 μM) with two of the compounds (Z56 and Z99) selectively killing BRCA-mutant cells and inhibiting cellular activities of RAD52 at micromolar inhibitor concentrations. While Z56 had no effect on the ssDNA-binding protein RPA and was toxic to BRCA-mutant cells only, Z99 inhibited both proteins and displayed toxicity towards BRCA-complemented cells. Optimization of the Z99 scaffold resulted in a set of more powerful and selective inhibitors (IC<sub>50</sub> ∼1.3-8 μM), which were only toxic to BRCA-mutant cells. RAD52 complexation by Z56, Z99 and its more specific derivatives provide a roadmap for next generation of cancer therapeutics.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/1c/4d/zcad018.PMC10150327.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9797966","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Deletion of the mRNA stability factor ELAVL1 (HuR) in pancreatic cancer cells disrupts the tumor microenvironment integrity. 胰腺癌症细胞中mRNA稳定因子ELAVL1(HuR)的缺失破坏了肿瘤微环境的完整性。
Pub Date : 2023-04-19 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad016
Grace A McCarthy, Roberto Di Niro, Jennifer M Finan, Aditi Jain, Yifei Guo, Cory R Wyatt, Alexander R Guimaraes, Trent A Waugh, Dove Keith, Terry K Morgan, Rosalie C Sears, Jonathan R Brody

Stromal cells promote extensive fibrosis in pancreatic ductal adenocarcinoma (PDAC), which is associated with poor prognosis and therapeutic resistance. We report here for the first time that loss of the RNA-binding protein human antigen R (HuR, ELAVL1) in PDAC cells leads to reprogramming of the tumor microenvironment. In multiple in vivo models, CRISPR deletion of ELAVL1 in PDAC cells resulted in a decrease of collagen deposition, accompanied by a decrease of stromal markers (i.e. podoplanin, α-smooth muscle actin, desmin). RNA-sequencing data showed that HuR plays a role in cell-cell communication. Accordingly, cytokine arrays identified that HuR regulates the secretion of signaling molecules involved in stromal activation and extracellular matrix organization [i.e. platelet-derived growth factor AA (PDGFAA) and pentraxin 3]. Ribonucleoprotein immunoprecipitation analysis and transcription inhibition studies validated PDGFA mRNA as a novel HuR target. These data suggest that tumor-intrinsic HuR supports extrinsic activation of the stroma to produce collagen and desmoplasia through regulating signaling molecules (e.g. PDGFAA). HuR-deficient PDAC in vivo tumors with an altered tumor microenvironment are more sensitive to the standard of care gemcitabine, as compared to HuR-proficient tumors. Taken together, we identified a novel role of tumor-intrinsic HuR in its ability to modify the surrounding tumor microenvironment and regulate PDGFAA.

基质细胞促进胰腺导管腺癌(PDAC)的广泛纤维化,这与不良预后和治疗耐药性有关。我们在这里首次报道了PDAC细胞中RNA结合蛋白人类抗原R(HuR,ELAVL1)的缺失导致肿瘤微环境的重新编程。在多种体内模型中,PDAC细胞中ELAVL1的CRISPR缺失导致胶原沉积减少,同时基质标记物(即足平面蛋白、α-平滑肌肌动蛋白、结蛋白)减少。RNA测序数据显示HuR在细胞间通讯中发挥作用。因此,细胞因子阵列鉴定出HuR调节参与基质激活和细胞外基质组织的信号分子的分泌[即血小板衍生生长因子AA(PDGFAA)和五唑菌素3]。核糖核蛋白免疫沉淀分析和转录抑制研究证实PDGFA mRNA是一种新的HuR靶点。这些数据表明,肿瘤内在的HuR支持基质的外在激活,通过调节信号分子(如PDGFAA)产生胶原和结缔组织增生。与精通HuR的肿瘤相比,肿瘤微环境改变的HuR缺陷型PDAC体内肿瘤对吉西他滨的标准护理更敏感。总之,我们确定了肿瘤内在HuR在其改变周围肿瘤微环境和调节PDGFAA的能力中的新作用。
{"title":"Deletion of the mRNA stability factor <i>ELAVL1</i> (HuR) in pancreatic cancer cells disrupts the tumor microenvironment integrity.","authors":"Grace A McCarthy, Roberto Di Niro, Jennifer M Finan, Aditi Jain, Yifei Guo, Cory R Wyatt, Alexander R Guimaraes, Trent A Waugh, Dove Keith, Terry K Morgan, Rosalie C Sears, Jonathan R Brody","doi":"10.1093/narcan/zcad016","DOIUrl":"10.1093/narcan/zcad016","url":null,"abstract":"<p><p>Stromal cells promote extensive fibrosis in pancreatic ductal adenocarcinoma (PDAC), which is associated with poor prognosis and therapeutic resistance. We report here for the first time that loss of the RNA-binding protein human antigen R (HuR, <i>ELAVL1</i>) in PDAC cells leads to reprogramming of the tumor microenvironment. In multiple <i>in vivo</i> models, CRISPR deletion of <i>ELAVL1</i> in PDAC cells resulted in a decrease of collagen deposition, accompanied by a decrease of stromal markers (i.e. podoplanin, α-smooth muscle actin, desmin). RNA-sequencing data showed that HuR plays a role in cell-cell communication. Accordingly, cytokine arrays identified that HuR regulates the secretion of signaling molecules involved in stromal activation and extracellular matrix organization [i.e. platelet-derived growth factor AA (PDGFAA) and pentraxin 3]. Ribonucleoprotein immunoprecipitation analysis and transcription inhibition studies validated <i>PDGFA</i> mRNA as a novel HuR target. These data suggest that tumor-intrinsic HuR supports extrinsic activation of the stroma to produce collagen and desmoplasia through regulating signaling molecules (e.g. PDGFAA). HuR-deficient PDAC <i>in vivo</i> tumors with an altered tumor microenvironment are more sensitive to the standard of care gemcitabine, as compared to HuR-proficient tumors. Taken together, we identified a novel role of tumor-intrinsic HuR in its ability to modify the surrounding tumor microenvironment and regulate PDGFAA.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/a9/5c/zcad016.PMC10113877.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9387874","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular origins of mutational spectra produced by the environmental carcinogen N-nitrosodimethylamine and SN1 chemotherapeutic agents. 环境致癌物质 N-亚硝基二甲胺和 SN1 化疗药物产生突变谱的分子起源。
Pub Date : 2023-03-27 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad015
Amanda L Armijo, Pennapa Thongararm, Bogdan I Fedeles, Judy Yau, Jennifer E Kay, Joshua J Corrigan, Marisa Chancharoen, Supawadee Chawanthayatham, Leona D Samson, Sebastian E Carrasco, Bevin P Engelward, James G Fox, Robert G Croy, John M Essigmann

DNA-methylating environmental carcinogens such as N-nitrosodimethylamine (NDMA) and certain alkylators used in chemotherapy form O 6-methylguanine (m6G) as a functionally critical intermediate. NDMA is a multi-organ carcinogen found in contaminated water, polluted air, preserved foods, tobacco products, and many pharmaceuticals. Only ten weeks after exposure to NDMA, neonatally-treated mice experienced elevated mutation frequencies in liver, lung and kidney of ∼35-fold, 4-fold and 2-fold, respectively. High-resolution mutational spectra (HRMS) of liver and lung revealed distinctive patterns dominated by GC→AT mutations in 5'-Pu-G-3' contexts, very similar to human COSMIC mutational signature SBS11. Commonly associated with alkylation damage, SBS11 appears in cancers treated with the DNA alkylator temozolomide (TMZ). When cells derived from the mice were treated with TMZ, N-methyl-N-nitrosourea, and streptozotocin (two other therapeutic methylating agents), all displayed NDMA-like HRMS, indicating mechanistically convergent mutational processes. The role of m6G in shaping the mutational spectrum of NDMA was probed by removing MGMT, the main cellular defense against m6G. MGMT-deficient mice displayed a strikingly enhanced mutant frequency, but identical HRMS, indicating that the mutational properties of these alkylators is likely owed to sequence-specific DNA binding. In sum, the HRMS of m6G-forming agents constitute an early-onset biomarker of exposure to DNA methylating carcinogens and drugs.

DNA 甲基化环境致癌物,如 N-亚硝基二甲胺(NDMA)和化疗中使用的某些烷化剂会形成 O-6-甲基鸟嘌呤(m6G),作为功能上的关键中间体。NDMA 是一种多器官致癌物质,存在于受污染的水、受污染的空气、腌制食品、烟草制品和许多药品中。接触 NDMA 仅十周后,新生小鼠肝脏、肺部和肾脏的突变频率分别升高了 35 倍、4 倍和 2 倍。肝脏和肺脏的高分辨率突变光谱(HRMS)显示,5'-Pu-G-3'背景下的GC→AT突变占主导地位,与人类COSMIC突变特征SBS11非常相似。SBS11通常与烷基化损伤有关,出现在使用DNA烷基化剂替莫唑胺(TMZ)治疗的癌症中。当小鼠的细胞接受 TMZ、N-甲基-N-亚硝基脲和链脲佐菌素(另外两种甲基化治疗剂)治疗时,所有细胞都显示出类似 NDMA 的 HRMS,这表明突变过程在机制上是趋同的。通过移除 MGMT(细胞对抗 m6G 的主要防御机制),探究了 m6G 在形成 NDMA 突变谱中的作用。缺失 MGMT 的小鼠突变频率显著增加,但 HRMS 却完全相同,这表明这些烷化剂的突变特性可能是由于序列特异性 DNA 结合所致。总之,m6G 形成剂的 HRMS 是暴露于 DNA 甲基化致癌物和药物的早期生物标志物。
{"title":"Molecular origins of mutational spectra produced by the environmental carcinogen <i>N</i>-nitrosodimethylamine and S<sub>N</sub>1 chemotherapeutic agents.","authors":"Amanda L Armijo, Pennapa Thongararm, Bogdan I Fedeles, Judy Yau, Jennifer E Kay, Joshua J Corrigan, Marisa Chancharoen, Supawadee Chawanthayatham, Leona D Samson, Sebastian E Carrasco, Bevin P Engelward, James G Fox, Robert G Croy, John M Essigmann","doi":"10.1093/narcan/zcad015","DOIUrl":"10.1093/narcan/zcad015","url":null,"abstract":"<p><p>DNA-methylating environmental carcinogens such as <i>N</i>-nitrosodimethylamine (NDMA) and certain alkylators used in chemotherapy form <i>O</i> <sup>6</sup>-methylguanine (m6G) as a functionally critical intermediate. NDMA is a multi-organ carcinogen found in contaminated water, polluted air, preserved foods, tobacco products, and many pharmaceuticals. Only ten weeks after exposure to NDMA, neonatally-treated mice experienced elevated mutation frequencies in liver, lung and kidney of ∼35-fold, 4-fold and 2-fold, respectively. High-resolution mutational spectra (HRMS) of liver and lung revealed distinctive patterns dominated by GC→AT mutations in 5'-Pu-G-3' contexts, very similar to human COSMIC mutational signature SBS11. Commonly associated with alkylation damage, SBS11 appears in cancers treated with the DNA alkylator temozolomide (TMZ). When cells derived from the mice were treated with TMZ, <i>N</i>-methyl-<i>N</i>-nitrosourea, and streptozotocin (two other therapeutic methylating agents), all displayed NDMA-like HRMS, indicating mechanistically convergent mutational processes. The role of m6G in shaping the mutational spectrum of NDMA was probed by removing MGMT, the main cellular defense against m6G. MGMT-deficient mice displayed a strikingly enhanced mutant frequency, but identical HRMS, indicating that the mutational properties of these alkylators is likely owed to sequence-specific DNA binding. In sum, the HRMS of m6G-forming agents constitute an early-onset biomarker of exposure to DNA methylating carcinogens and drugs.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-03-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10041537/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9743909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The hereditary N363K POLE exonuclease mutant extends PPAP tumor spectrum to glioblastomas by causing DNA damage and aneuploidy in addition to increased mismatch mutagenicity. 遗传性 N363K POLE 外切酶突变体除了增加错配诱变性外,还造成 DNA 损伤和非整倍体,从而将 PPAP 的肿瘤谱扩展到胶质母细胞瘤。
Pub Date : 2023-03-11 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad011
Guillaume Labrousse, Pierre Vande Perre, Genis Parra, Marion Jaffrelot, Laura Leroy, Frederic Chibon, Frederic Escudie, Janick Selves, Jean-Sebastien Hoffmann, Rosine Guimbaud, Malik Lutzmann

The exonuclease domain of DNA polymerases epsilon's catalytic subunit (POLE) removes misincorporated nucleotides, called proofreading. POLE-exonuclease mutations cause colorectal- and endometrial cancers with an extreme burden of single nucleotide substitutions. We recently reported that particularly the hereditary POLE exonuclease mutation N363K predisposes in addition to aggressive giant cell glioblastomas. We knocked-in this mutation homozygously into human cell lines and compared its properties to knock-ins of the likewise hereditary POLE L424V mutation and to a complete proofreading-inactivating mutation (exo-null). We found that N363K cells have higher mutation rates as both L424V- or exo-null mutant cells. In contrast to L424V cells, N363K cells expose a growth defect, replication stress and DNA damage. In non-transformed cells, these burdens lead to aneuploidy but macroscopically normal nuclei. In contrast, transformed N363K cells phenocopy the enlarged and disorganized nuclei of giant cell glioblastomas. Taken together, our data characterize a POLE exonuclease domain mutant that not only causes single nucleotide hypermutation, but in addition DNA damage and chromosome instability, leading to an extended tumor spectrum. Our results expand the understanding of the polymerase exonuclease domain and suggest that an assessment of both the mutational potential and the genetic instability might refine classification and treatment of POLE-mutated tumors.

DNA 聚合酶epsilon催化亚基(POLE)的外切酶结构域能清除误入的核苷酸,称为校对。POLE-exonuclease 基因突变导致的结直肠癌和子宫内膜癌中,单核苷酸置换的比例极高。我们最近报告说,遗传性 POLE 外切酶突变 N363K 除了容易导致侵袭性巨细胞胶质母细胞瘤外,还容易导致遗传性 POLE 外切酶突变 N363K。我们在人类细胞系中同源敲入了这一突变,并将其特性与同样具有遗传性的 POLE L424V 突变的敲入和完全校对失活突变(exo-null)进行了比较。我们发现,N363K 细胞的突变率高于 L424V 突变细胞或 exo-null 突变细胞。与 L424V 细胞相比,N363K 细胞暴露出生长缺陷、复制压力和 DNA 损伤。在非转化细胞中,这些负担会导致非整倍体,但细胞核宏观上正常。相反,转化的 N363K 细胞则表现出巨细胞胶质母细胞瘤细胞核的增大和紊乱。综上所述,我们的数据描述了一种 POLE 外切酶结构域突变体的特征,它不仅会引起单核苷酸高突变,还会造成 DNA 损伤和染色体不稳定,从而导致肿瘤范围扩大。我们的研究结果拓展了人们对聚合酶外切酶结构域的认识,并表明对突变潜能和遗传不稳定性的评估可能会完善POLE突变肿瘤的分类和治疗。
{"title":"The hereditary N363K POLE exonuclease mutant extends PPAP tumor spectrum to glioblastomas by causing DNA damage and aneuploidy in addition to increased mismatch mutagenicity.","authors":"Guillaume Labrousse, Pierre Vande Perre, Genis Parra, Marion Jaffrelot, Laura Leroy, Frederic Chibon, Frederic Escudie, Janick Selves, Jean-Sebastien Hoffmann, Rosine Guimbaud, Malik Lutzmann","doi":"10.1093/narcan/zcad011","DOIUrl":"10.1093/narcan/zcad011","url":null,"abstract":"<p><p>The exonuclease domain of DNA polymerases epsilon's catalytic subunit (POLE) removes misincorporated nucleotides, called proofreading. POLE-exonuclease mutations cause colorectal- and endometrial cancers with an extreme burden of single nucleotide substitutions. We recently reported that particularly the hereditary POLE exonuclease mutation N363K predisposes in addition to aggressive giant cell glioblastomas. We knocked-in this mutation homozygously into human cell lines and compared its properties to knock-ins of the likewise hereditary POLE L424V mutation and to a complete proofreading-inactivating mutation (exo-null). We found that N363K cells have higher mutation rates as both L424V- or exo-null mutant cells. In contrast to L424V cells, N363K cells expose a growth defect, replication stress and DNA damage. In non-transformed cells, these burdens lead to aneuploidy but macroscopically normal nuclei. In contrast, transformed N363K cells phenocopy the enlarged and disorganized nuclei of giant cell glioblastomas. Taken together, our data characterize a POLE exonuclease domain mutant that not only causes single nucleotide hypermutation, but in addition DNA damage and chromosome instability, leading to an extended tumor spectrum. Our results expand the understanding of the polymerase exonuclease domain and suggest that an assessment of both the mutational potential and the genetic instability might refine classification and treatment of POLE-mutated tumors.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-03-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/fd/95/zcad011.PMC10006997.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9168828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ICI efficacy information portal: a knowledgebase for responder prediction to immune checkpoint inhibitors. ICI 疗效信息门户:免疫检查点抑制剂应答者预测知识库。
Pub Date : 2023-03-03 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad012
Jiamin Chen, Daniel Rebibo, Jianquan Cao, Simon Yat-Man Mok, Neel Patel, Po-Cheng Tseng, Zhenghao Zhang, Kevin Y Yip

Immune checkpoint inhibitors (ICIs) have led to durable responses in cancer patients, yet their efficacy varies significantly across cancer types and patients. To stratify patients based on their potential clinical benefits, there have been substantial research efforts in identifying biomarkers and computational models that can predict the efficacy of ICIs, and it has become difficult to keep track of all of them. It is also difficult to compare findings of different studies since they involve different cancer types, ICIs, and various other details. To make it easy to access the latest information about ICI efficacy, we have developed a knowledgebase and a corresponding web-based portal (https://iciefficacy.org/). Our knowledgebase systematically records information about latest publications related to ICI efficacy, predictors proposed, and datasets used to test them. All information recorded is checked carefully by a manual curation process. The web-based portal provides functions to browse, search, filter, and sort the information. Digests of method details are provided based on the original descriptions in the publications. Evaluation results of the effectiveness of the predictors reported in the publications are summarized for quick overviews. Overall, our resource provides centralized access to the burst of information produced by the vibrant research on ICI efficacy.

免疫检查点抑制剂(ICIs)为癌症患者带来了持久的疗效,但其疗效在不同癌症类型和患者之间存在很大差异。为了根据潜在的临床疗效对患者进行分层,研究人员一直在努力寻找可以预测 ICIs 疗效的生物标记物和计算模型,但要跟踪所有这些标记物和模型已变得十分困难。此外,由于不同的研究涉及不同的癌症类型、ICIs 及其他各种细节,因此也很难对不同研究的结果进行比较。为了方便获取有关 ICI 疗效的最新信息,我们开发了一个知识库和相应的网络门户网站 (https://iciefficacy.org/)。我们的知识库系统地记录了与 ICI 疗效相关的最新出版物、提出的预测指标以及用于测试这些指标的数据集等信息。所有记录的信息都经过人工整理过程的仔细检查。基于网络的门户网站提供浏览、搜索、过滤和排序信息的功能。根据出版物中的原始描述,提供了方法细节的摘要。对出版物中报告的预测因子有效性的评估结果进行了总结,以便快速浏览。总之,我们的资源提供了集中访问 ICI 功效研究产生的大量信息的途径。
{"title":"ICI efficacy information portal: a knowledgebase for responder prediction to immune checkpoint inhibitors.","authors":"Jiamin Chen, Daniel Rebibo, Jianquan Cao, Simon Yat-Man Mok, Neel Patel, Po-Cheng Tseng, Zhenghao Zhang, Kevin Y Yip","doi":"10.1093/narcan/zcad012","DOIUrl":"10.1093/narcan/zcad012","url":null,"abstract":"<p><p>Immune checkpoint inhibitors (ICIs) have led to durable responses in cancer patients, yet their efficacy varies significantly across cancer types and patients. To stratify patients based on their potential clinical benefits, there have been substantial research efforts in identifying biomarkers and computational models that can predict the efficacy of ICIs, and it has become difficult to keep track of all of them. It is also difficult to compare findings of different studies since they involve different cancer types, ICIs, and various other details. To make it easy to access the latest information about ICI efficacy, we have developed a knowledgebase and a corresponding web-based portal (https://iciefficacy.org/). Our knowledgebase systematically records information about latest publications related to ICI efficacy, predictors proposed, and datasets used to test them. All information recorded is checked carefully by a manual curation process. The web-based portal provides functions to browse, search, filter, and sort the information. Digests of method details are provided based on the original descriptions in the publications. Evaluation results of the effectiveness of the predictors reported in the publications are summarized for quick overviews. Overall, our resource provides centralized access to the burst of information produced by the vibrant research on ICI efficacy.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9984987/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9612060","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Recent advancements in the structural biology of human telomerase and their implications for improved design of cancer therapeutics. 人类端粒酶结构生物学的最新进展及其对癌症治疗方法改进设计的意义。
Pub Date : 2023-03-03 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad010
Griffin A Welfer, Bret D Freudenthal

Telomerase is a specialized reverse transcriptase that synthesizes telomeric repeats at the ends of linear chromosomes. Telomerase is transiently expressed in germ and stem cells, but nearly all somatic cells silence it after differentiating. However, the vast majority of cancer cells reactivate and constitutively express telomerase to maintain replicative immortality. Because of this, telomerase has remained a promising broad-spectrum chemotherapeutic target for over 30 years. However, various challenges associated with obtaining high-resolution structural data for telomerase have limited the development of rationally designed structure-based therapeutics. Various techniques and model systems have been utilized to advance our understanding of the structural biology of telomerase. In particular, multiple high-resolution cryogenic electron microscopy (cryo-EM) structures published within the past few years have revealed new components of the telomerase complex with near atomic resolution structural models. Additionally, these structures have provided details for how telomerase is recruited to telomeres and its mechanism of telomere synthesis. With these new pieces of evidence, and the promising outlook for future refinements to our current models, the possibility of telomerase specific chemotherapeutics is becoming more tangible than ever. This review summarizes these recent advancements and outlines outstanding questions in the field.

端粒酶是一种专门的逆转录酶,在线性染色体末端合成端粒重复序列。端粒酶在生殖细胞和干细胞中瞬时表达,但几乎所有体细胞在分化后都会使其沉默。然而,绝大多数癌症细胞重新激活并组成性表达端粒酶,以维持复制永生。正因为如此,30多年来,端粒酶一直是一种很有前途的广谱化疗靶点。然而,与获得端粒酶高分辨率结构数据相关的各种挑战限制了合理设计的基于结构的治疗方法的发展。各种技术和模型系统已被用于促进我们对端粒酶结构生物学的理解。特别是,在过去几年中发表的多个高分辨率低温电子显微镜(cryo-EM)结构已经通过近原子分辨率的结构模型揭示了端粒酶复合物的新成分。此外,这些结构为端粒酶如何被募集到端粒及其端粒合成机制提供了细节。有了这些新的证据,以及对我们当前模型未来改进的前景,端粒酶特异性化疗的可能性比以往任何时候都更加明显。这篇综述总结了这些最新进展,并概述了该领域悬而未决的问题。
{"title":"Recent advancements in the structural biology of human telomerase and their implications for improved design of cancer therapeutics.","authors":"Griffin A Welfer, Bret D Freudenthal","doi":"10.1093/narcan/zcad010","DOIUrl":"10.1093/narcan/zcad010","url":null,"abstract":"<p><p>Telomerase is a specialized reverse transcriptase that synthesizes telomeric repeats at the ends of linear chromosomes. Telomerase is transiently expressed in germ and stem cells, but nearly all somatic cells silence it after differentiating. However, the vast majority of cancer cells reactivate and constitutively express telomerase to maintain replicative immortality. Because of this, telomerase has remained a promising broad-spectrum chemotherapeutic target for over 30 years. However, various challenges associated with obtaining high-resolution structural data for telomerase have limited the development of rationally designed structure-based therapeutics. Various techniques and model systems have been utilized to advance our understanding of the structural biology of telomerase. In particular, multiple high-resolution cryogenic electron microscopy (cryo-EM) structures published within the past few years have revealed new components of the telomerase complex with near atomic resolution structural models. Additionally, these structures have provided details for how telomerase is recruited to telomeres and its mechanism of telomere synthesis. With these new pieces of evidence, and the promising outlook for future refinements to our current models, the possibility of telomerase specific chemotherapeutics is becoming more tangible than ever. This review summarizes these recent advancements and outlines outstanding questions in the field.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9984990/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9112498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
R-loops, type I topoisomerases and cancer. R环、I型拓扑异构酶与癌症
Pub Date : 2023-03-03 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad013
Sourav Saha, Yves Pommier

R-loops are abundant and dynamic structures ubiquitously present in human cells both in the nuclear and mitochondrial genomes. They form in cis in the wake of transcription complexes and in trans apart from transcription complexes. In this review, we focus on the relationship between R-loops and topoisomerases, and cancer genomics and therapies. We summarize the topological parameters associated with the formation and resolution of R-loops, which absorb and release high levels of genomic negative supercoiling (Sc-). We review the deleterious consequences of excessive R-loops and rationalize how human type IA (TOP3B) and type IB (TOP1) topoisomerases regulate and resolve R-loops in coordination with helicase and RNase H enzymes. We also review the drugs (topoisomerase inhibitors, splicing inhibitors, G4 stabilizing ligands) and cancer predisposing genes (BRCA1/2, transcription, and splicing genes) known to induce R-loops, and whether stabilizing R-loops and thereby inducing genomic damage can be viewed as a strategy for cancer treatment.

在人类细胞的核基因组和线粒体基因组中,R 环是一种丰富的动态结构。它们顺式形成于转录复合体之后,反式形成于转录复合体之外。在这篇综述中,我们将重点讨论 R 环和拓扑异构酶与癌症基因组学和疗法之间的关系。我们总结了与 R 环的形成和解析相关的拓扑参数,R 环吸收并释放大量基因组负超螺旋(Sc-)。我们回顾了过多 R 环的有害后果,并合理解释了人类 IA 型(TOP3B)和 IB 型(TOP1)拓扑异构酶如何与螺旋酶和 RNase H 酶协调调节和解决 R 环。我们还回顾了已知可诱导 R 环的药物(拓扑异构酶抑制剂、剪接抑制剂、G4 稳定配体)和癌症易感基因(BRCA1/2、转录和剪接基因),以及是否可将稳定 R 环从而诱导基因组损伤视为一种癌症治疗策略。
{"title":"R-loops, type I topoisomerases and cancer.","authors":"Sourav Saha, Yves Pommier","doi":"10.1093/narcan/zcad013","DOIUrl":"10.1093/narcan/zcad013","url":null,"abstract":"<p><p>R-loops are abundant and dynamic structures ubiquitously present in human cells both in the nuclear and mitochondrial genomes. They form in <i>cis</i> in the wake of transcription complexes and in <i>trans</i> apart from transcription complexes. In this review, we focus on the relationship between R-loops and topoisomerases, and cancer genomics and therapies. We summarize the topological parameters associated with the formation and resolution of R-loops, which absorb and release high levels of genomic negative supercoiling (Sc-). We review the deleterious consequences of excessive R-loops and rationalize how human type IA (TOP3B) and type IB (TOP1) topoisomerases regulate and resolve R-loops in coordination with helicase and RNase H enzymes. We also review the drugs (topoisomerase inhibitors, splicing inhibitors, G4 stabilizing ligands) and cancer predisposing genes (BRCA1/2, transcription, and splicing genes) known to induce R-loops, and whether stabilizing R-loops and thereby inducing genomic damage can be viewed as a strategy for cancer treatment.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-03-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9984992/pdf/zcad013.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10048600","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
NAR Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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