首页 > 最新文献

NAR Cancer最新文献

英文 中文
Synthetic lethal interaction between WEE1 and PKMYT1 is a target for multiple low-dose treatment of high-grade serous ovarian carcinoma. WEE1和PKMYT1之间的合成致死相互作用是高级别浆液性卵巢癌多次低剂量治疗的靶点。
Pub Date : 2023-06-13 eCollection Date: 2023-09-01 DOI: 10.1093/narcan/zcad029
Jan Benada, Daria Bulanova, Violette Azzoni, Valdemaras Petrosius, Saba Ghazanfar, Krister Wennerberg, Claus Storgaard Sørensen

Ovarian cancer is driven by genetic alterations that necessitate protective DNA damage and replication stress responses through cell cycle control and genome maintenance. This creates specific vulnerabilities that may be exploited therapeutically. WEE1 kinase is a key cell cycle control kinase, and it has emerged as a promising cancer therapy target. However, adverse effects have limited its clinical progress, especially when tested in combination with chemotherapies. A strong genetic interaction between WEE1 and PKMYT1 led us to hypothesize that a multiple low-dose approach utilizing joint WEE1 and PKMYT1 inhibition would allow exploitation of the synthetic lethality. We found that the combination of WEE1 and PKMYT1 inhibition exhibited synergistic effects in eradicating ovarian cancer cells and organoid models at a low dose. The WEE1 and PKMYT1 inhibition synergistically promoted CDK activation. Furthermore, the combined treatment exacerbated DNA replication stress and replication catastrophe, leading to increase of the genomic instability and inflammatory STAT1 signalling activation. These findings suggest a new multiple low-dose approach to harness the potency of WEE1 inhibition through the synthetic lethal interaction with PKMYT1 that may contribute to the development of new treatments for ovarian cancer.

卵巢癌是由基因改变驱动的,这种改变需要通过细胞周期控制和基因组维护来做出保护性 DNA 损伤和复制应激反应。这就造成了一些可被治疗利用的特殊弱点。WEE1 激酶是一种关键的细胞周期控制激酶,已成为一种很有前景的癌症治疗靶点。然而,不良反应限制了其临床进展,尤其是在与化疗药物联合使用时。WEE1和PKMYT1之间存在很强的遗传相互作用,这使我们假设,利用联合抑制WEE1和PKMYT1的多重低剂量方法可以利用合成致死性。我们发现,WEE1 和 PKMYT1 联合抑制在低剂量根除卵巢癌细胞和类器官模型方面表现出协同效应。WEE1和PKMYT1抑制剂协同促进CDK活化。此外,联合治疗会加剧 DNA 复制应激和复制灾难,导致基因组不稳定性增加和 STAT1 信号激活。这些研究结果表明,通过与PKMYT1的合成致死相互作用,一种新的多重低剂量方法可利用WEE1的抑制作用,这可能有助于开发治疗卵巢癌的新疗法。
{"title":"Synthetic lethal interaction between WEE1 and PKMYT1 is a target for multiple low-dose treatment of high-grade serous ovarian carcinoma.","authors":"Jan Benada, Daria Bulanova, Violette Azzoni, Valdemaras Petrosius, Saba Ghazanfar, Krister Wennerberg, Claus Storgaard Sørensen","doi":"10.1093/narcan/zcad029","DOIUrl":"10.1093/narcan/zcad029","url":null,"abstract":"<p><p>Ovarian cancer is driven by genetic alterations that necessitate protective DNA damage and replication stress responses through cell cycle control and genome maintenance. This creates specific vulnerabilities that may be exploited therapeutically. WEE1 kinase is a key cell cycle control kinase, and it has emerged as a promising cancer therapy target. However, adverse effects have limited its clinical progress, especially when tested in combination with chemotherapies. A strong genetic interaction between WEE1 and PKMYT1 led us to hypothesize that a multiple low-dose approach utilizing joint WEE1 and PKMYT1 inhibition would allow exploitation of the synthetic lethality. We found that the combination of WEE1 and PKMYT1 inhibition exhibited synergistic effects in eradicating ovarian cancer cells and organoid models at a low dose. The WEE1 and PKMYT1 inhibition synergistically promoted CDK activation. Furthermore, the combined treatment exacerbated DNA replication stress and replication catastrophe, leading to increase of the genomic instability and inflammatory STAT1 signalling activation. These findings suggest a new multiple low-dose approach to harness the potency of WEE1 inhibition through the synthetic lethal interaction with PKMYT1 that may contribute to the development of new treatments for ovarian cancer.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10262308/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9657339","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
DNA mismatch repair in cancer immunotherapy. 癌症免疫疗法中的 DNA 错配修复。
Pub Date : 2023-06-13 eCollection Date: 2023-09-01 DOI: 10.1093/narcan/zcad031
Junhong Guan, Guo-Min Li

Tumors defective in DNA mismatch repair (dMMR) exhibit microsatellite instability (MSI). Currently, patients with dMMR tumors are benefitted from anti-PD-1/PDL1-based immune checkpoint inhibitor (ICI) therapy. Over the past several years, great progress has been made in understanding the mechanisms by which dMMR tumors respond to ICI, including the identification of mutator phenotype-generated neoantigens, cytosolic DNA-mediated activation of the cGAS-STING pathway, type-I interferon signaling and high tumor-infiltration of lymphocytes in dMMR tumors. Although ICI therapy shows great clinical benefits, ∼50% of dMMR tumors are eventually not responsive. Here we review the discovery, development and molecular basis of dMMR-mediated immunotherapy, as well as tumor resistant problems and potential therapeutic interventions to overcome the resistance.

DNA 错配修复(dMMR)缺陷肿瘤表现出微卫星不稳定性(MSI)。目前,dMMR肿瘤患者可从基于抗PD-1/PDL1的免疫检查点抑制剂(ICI)疗法中获益。在过去几年中,人们在了解 dMMR 肿瘤对 ICI 的反应机制方面取得了重大进展,包括确定了突变体表型产生的新抗原、细胞膜 DNA 介导的 cGAS-STING 通路激活、I 型干扰素信号转导以及 dMMR 肿瘤中淋巴细胞的高肿瘤浸润。尽管 ICI 疗法显示出巨大的临床疗效,但仍有 50% 的 dMMR 肿瘤最终没有反应。在此,我们回顾了dMMR介导的免疫疗法的发现、发展和分子基础,以及肿瘤耐药性问题和克服耐药性的潜在治疗干预措施。
{"title":"DNA mismatch repair in cancer immunotherapy.","authors":"Junhong Guan, Guo-Min Li","doi":"10.1093/narcan/zcad031","DOIUrl":"10.1093/narcan/zcad031","url":null,"abstract":"<p><p>Tumors defective in DNA mismatch repair (dMMR) exhibit microsatellite instability (MSI). Currently, patients with dMMR tumors are benefitted from anti-PD-1/PDL1-based immune checkpoint inhibitor (ICI) therapy. Over the past several years, great progress has been made in understanding the mechanisms by which dMMR tumors respond to ICI, including the identification of mutator phenotype-generated neoantigens, cytosolic DNA-mediated activation of the cGAS-STING pathway, type-I interferon signaling and high tumor-infiltration of lymphocytes in dMMR tumors. Although ICI therapy shows great clinical benefits, ∼50% of dMMR tumors are eventually not responsive. Here we review the discovery, development and molecular basis of dMMR-mediated immunotherapy, as well as tumor resistant problems and potential therapeutic interventions to overcome the resistance.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/5f/a6/zcad031.PMC10262306.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9657336","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
TP53 germline pathogenic variants in modern humans were likely originated during recent human history. 现代人中的 TP53 基因致病变体很可能起源于近代人类历史。
Pub Date : 2023-06-09 eCollection Date: 2023-09-01 DOI: 10.1093/narcan/zcad025
Si Hoi Kou, Jiaheng Li, Benjamin Tam, Huijun Lei, Bojin Zhao, Fengxia Xiao, San Ming Wang

TP53 is crucial for maintaining genome stability and preventing oncogenesis. Germline pathogenic variation in TP53 damages its function, causing genome instability and increased cancer risk. Despite extensive study in TP53, the evolutionary origin of the human TP53 germline pathogenic variants remains largely unclear. In this study, we applied phylogenetic and archaeological approaches to identify the evolutionary origin of TP53 germline pathogenic variants in modern humans. In the phylogenic analysis, we searched 406 human TP53 germline pathogenic variants in 99 vertebrates distributed in eight clades of Primate, Euarchontoglires, Laurasiatheria, Afrotheria, Mammal, Aves, Sarcopterygii and Fish, but we observed no direct evidence for the cross-species conservation as the origin; in the archaeological analysis, we searched the variants in 5031 ancient human genomes dated between 45045 and 100 years before present, and identified 45 pathogenic variants in 62 ancient humans dated mostly within the last 8000 years; we also identified 6 pathogenic variants in 3 Neanderthals dated 44000 to 38515 years before present and 1 Denisovan dated 158 550 years before present. Our study reveals that TP53 germline pathogenic variants in modern humans were likely originated in recent human history and partially inherited from the extinct Neanderthals and Denisovans.

TP53 对维持基因组稳定性和防止肿瘤发生至关重要。TP53 的种系致病变异会破坏其功能,导致基因组不稳定和癌症风险增加。尽管对 TP53 进行了广泛研究,但人类 TP53 种系致病变异的进化起源在很大程度上仍不清楚。在这项研究中,我们应用系统发生学和考古学方法来确定现代人类 TP53 生殖系致病变异体的进化起源。在系统发育分析中,我们搜索了分布在灵长类、始祖鸟类、月鸟类、非洲鸟类、哺乳类、鸟类、猿猴类和鱼类八个支系的 99 种脊椎动物中的 406 个人类 TP53 种系致病变体,但没有观察到跨物种保护作为起源的直接证据;在考古分析中,我们搜索了5031个古人类基因组中的变体,其年代在距今45045年到100年之间,在62个古人类基因组中发现了45个致病变体,其年代大多在距今8000年之内;我们还在3个尼安德特人基因组中发现了6个致病变体,其年代在距今44000年到38515年之间,在1个丹尼索瓦人基因组中发现了6个致病变体,其年代在距今158 550年之前。我们的研究表明,现代人的 TP53 生殖系致病变体很可能起源于近代人类历史,部分遗传自已经灭绝的尼安德特人和丹尼索瓦人。
{"title":"<i>TP53</i> germline pathogenic variants in modern humans were likely originated during recent human history.","authors":"Si Hoi Kou, Jiaheng Li, Benjamin Tam, Huijun Lei, Bojin Zhao, Fengxia Xiao, San Ming Wang","doi":"10.1093/narcan/zcad025","DOIUrl":"10.1093/narcan/zcad025","url":null,"abstract":"<p><p><i>TP53</i> is crucial for maintaining genome stability and preventing oncogenesis. Germline pathogenic variation in <i>TP53</i> damages its function, causing genome instability and increased cancer risk. Despite extensive study in <i>TP53</i>, the evolutionary origin of the human <i>TP53</i> germline pathogenic variants remains largely unclear. In this study, we applied phylogenetic and archaeological approaches to identify the evolutionary origin of <i>TP53</i> germline pathogenic variants in modern humans. In the phylogenic analysis, we searched 406 human <i>TP53</i> germline pathogenic variants in 99 vertebrates distributed in eight clades of Primate, Euarchontoglires, Laurasiatheria, Afrotheria, Mammal, Aves, Sarcopterygii and Fish, but we observed no direct evidence for the cross-species conservation as the origin; in the archaeological analysis, we searched the variants in 5031 ancient human genomes dated between 45045 and 100 years before present, and identified 45 pathogenic variants in 62 ancient humans dated mostly within the last 8000 years; we also identified 6 pathogenic variants in 3 Neanderthals dated 44000 to 38515 years before present and 1 Denisovan dated 158 550 years before present. Our study reveals that <i>TP53</i> germline pathogenic variants in modern humans were likely originated in recent human history and partially inherited from the extinct Neanderthals and Denisovans.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/83/a4/zcad025.PMC10251638.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9620780","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
Discovering cryptic splice mutations in cancers via a deep neural network framework. 通过深度神经网络框架发现癌症中的隐剪接突变。
Pub Date : 2023-06-01 DOI: 10.1093/narcan/zcad014
Raphaël Teboul, Michalina Grabias, Jessica Zucman-Rossi, Eric Letouzé

Somatic mutations can disrupt splicing regulatory elements and have dramatic effects on cancer genes, yet the functional consequences of mutations located in extended splice regions is difficult to predict. Here, we use a deep neural network (SpliceAI) to characterize the landscape of splice-altering mutations in cancer. In our in-house series of 401 liver cancers, SpliceAI uncovers 1244 cryptic splice mutations, located outside essential splice sites, that validate at a high rate (66%) in matched RNA-seq data. We then extend the analysis to a large pan-cancer cohort of 17 714 tumors, revealing >100 000 cryptic splice mutations. Taking into account these mutations increases the power of driver gene discovery, revealing 126 new candidate driver genes. It also reveals new driver mutations in known cancer genes, doubling the frequency of splice alterations in tumor suppressor genes. Mutational signature analysis suggests mutational processes that could give rise preferentially to splice mutations in each cancer type, with an enrichment of signatures related to clock-like processes and DNA repair deficiency. Altogether, this work sheds light on the causes and impact of cryptic splice mutations in cancer, and highlights the power of deep learning approaches to better annotate the functional consequences of mutations in oncology.

体细胞突变可以破坏剪接调节元件并对癌症基因产生巨大影响,然而位于扩展剪接区域的突变的功能后果很难预测。在这里,我们使用深度神经网络(SpliceAI)来表征癌症中剪接改变突变的景观。在我们的401例肝癌的内部系列中,SpliceAI发现了1244个隐剪接突变,位于必要剪接位点之外,在匹配的RNA-seq数据中验证率很高(66%)。然后,我们将分析扩展到17714个肿瘤的大型泛癌症队列,揭示了>10万个隐剪接突变。考虑到这些突变增加了发现驱动基因的能力,揭示了126个新的候选驱动基因。它还揭示了已知癌症基因中新的驱动突变,使肿瘤抑制基因剪接改变的频率增加了一倍。突变特征分析表明,突变过程可能在每种癌症类型中优先引起剪接突变,与时钟样过程和DNA修复缺陷相关的特征丰富。总之,这项工作揭示了癌症中隐剪接突变的原因和影响,并强调了深度学习方法在更好地注释肿瘤突变的功能后果方面的力量。
{"title":"Discovering cryptic splice mutations in cancers via a deep neural network framework.","authors":"Raphaël Teboul,&nbsp;Michalina Grabias,&nbsp;Jessica Zucman-Rossi,&nbsp;Eric Letouzé","doi":"10.1093/narcan/zcad014","DOIUrl":"https://doi.org/10.1093/narcan/zcad014","url":null,"abstract":"<p><p>Somatic mutations can disrupt splicing regulatory elements and have dramatic effects on cancer genes, yet the functional consequences of mutations located in extended splice regions is difficult to predict. Here, we use a deep neural network (SpliceAI) to characterize the landscape of splice-altering mutations in cancer. In our in-house series of 401 liver cancers, SpliceAI uncovers 1244 cryptic splice mutations, located outside essential splice sites, that validate at a high rate (66%) in matched RNA-seq data. We then extend the analysis to a large pan-cancer cohort of 17 714 tumors, revealing >100 000 cryptic splice mutations. Taking into account these mutations increases the power of driver gene discovery, revealing 126 new candidate driver genes. It also reveals new driver mutations in known cancer genes, doubling the frequency of splice alterations in tumor suppressor genes. Mutational signature analysis suggests mutational processes that could give rise preferentially to splice mutations in each cancer type, with an enrichment of signatures related to clock-like processes and DNA repair deficiency. Altogether, this work sheds light on the causes and impact of cryptic splice mutations in cancer, and highlights the power of deep learning approaches to better annotate the functional consequences of mutations in oncology.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10015341/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9200572","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
Estrogen receptor alpha mutations regulate gene expression and cell growth in breast cancer through microRNAs. 雌激素受体α突变通过微RNA调控乳腺癌的基因表达和细胞生长。
Pub Date : 2023-06-01 DOI: 10.1093/narcan/zcad027
Spencer Arnesen, Jacob T Polaski, Zannel Blanchard, Kyle S Osborne, Alana L Welm, Ryan M O'Connell, Jason Gertz

Estrogen receptor α (ER) mutations occur in up to 30% of metastatic ER-positive breast cancers. Recent data has shown that ER mutations impact the expression of thousands of genes not typically regulated by wildtype ER. While the majority of these altered genes can be explained by constant activity of mutant ER or genomic changes such as altered ER binding and chromatin accessibility, as much as 33% remain unexplained, indicating the potential for post-transcriptional effects. Here, we explored the role of microRNAs in mutant ER-driven gene regulation and identified several microRNAs that are dysregulated in ER mutant cells. These differentially regulated microRNAs target a significant portion of mutant-specific genes involved in key cellular processes. When the activity of microRNAs is altered using mimics or inhibitors, significant changes are observed in gene expression and cellular proliferation related to mutant ER. An in-depth evaluation of miR-301b led us to discover an important role for PRKD3 in the proliferation of ER mutant cells. Our findings show that microRNAs contribute to mutant ER gene regulation and cellular effects in breast cancer cells.

雌激素受体α(ER)突变发生在高达30%的ER阳性转移性乳腺癌中。最新数据显示,ER突变会影响数千个基因的表达,而这些基因通常不受野生型ER的调控。虽然这些改变的基因大部分可以用突变ER的持续活性或基因组变化(如ER结合和染色质可及性的改变)来解释,但仍有多达33%的基因无法解释,这表明可能存在转录后效应。在这里,我们探索了微RNA在突变ER驱动的基因调控中的作用,并确定了几种在ER突变细胞中调控失调的微RNA。这些受到不同调控的 microRNA 靶向了很大一部分参与关键细胞过程的突变特异性基因。当使用模拟物或抑制剂改变微RNA的活性时,就能观察到与突变ER相关的基因表达和细胞增殖发生了显著变化。对miR-301b的深入评估使我们发现了PRKD3在ER突变细胞增殖中的重要作用。我们的研究结果表明,microRNA 对乳腺癌细胞中突变 ER 基因的调控和细胞效应做出了贡献。
{"title":"Estrogen receptor alpha mutations regulate gene expression and cell growth in breast cancer through microRNAs.","authors":"Spencer Arnesen, Jacob T Polaski, Zannel Blanchard, Kyle S Osborne, Alana L Welm, Ryan M O'Connell, Jason Gertz","doi":"10.1093/narcan/zcad027","DOIUrl":"10.1093/narcan/zcad027","url":null,"abstract":"<p><p>Estrogen receptor α (ER) mutations occur in up to 30% of metastatic ER-positive breast cancers. Recent data has shown that ER mutations impact the expression of thousands of genes not typically regulated by wildtype ER. While the majority of these altered genes can be explained by constant activity of mutant ER or genomic changes such as altered ER binding and chromatin accessibility, as much as 33% remain unexplained, indicating the potential for post-transcriptional effects. Here, we explored the role of microRNAs in mutant ER-driven gene regulation and identified several microRNAs that are dysregulated in ER mutant cells. These differentially regulated microRNAs target a significant portion of mutant-specific genes involved in key cellular processes. When the activity of microRNAs is altered using mimics or inhibitors, significant changes are observed in gene expression and cellular proliferation related to mutant ER. An in-depth evaluation of miR-301b led us to discover an important role for <i>PRKD3</i> in the proliferation of ER mutant cells. Our findings show that microRNAs contribute to mutant ER gene regulation and cellular effects in breast cancer cells.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10233889/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10257360","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
HiTAIC: hierarchical tumor artificial intelligence classifier traces tissue of origin and tumor type in primary and metastasized tumors using DNA methylation. HiTAIC:分级肿瘤人工智能分类器利用DNA甲基化追踪原发性和转移性肿瘤的起源组织和肿瘤类型。
Pub Date : 2023-06-01 DOI: 10.1093/narcan/zcad017
Ze Zhang, Yunrui Lu, Soroush Vosoughi, Joshua J Levy, Brock C Christensen, Lucas A Salas

Human cancers are heterogenous by their cell composition and origination site. Cancer metastasis generates the conundrum of the unknown origin of migrated tumor cells. Tracing tissue of origin and tumor type in primary and metastasized cancer is vital for clinical significance. DNA methylation alterations play a crucial role in carcinogenesis and mark cell fate differentiation, thus can be used to trace tumor tissue of origin. In this study, we employed a novel tumor-type-specific hierarchical model using genome-scale DNA methylation data to develop a multilayer perceptron model, HiTAIC, to trace tissue of origin and tumor type in 27 cancers from 23 tissue sites in data from 7735 tumors with high resolution, accuracy, and specificity. In tracing primary cancer origin, HiTAIC accuracy was 99% in the test set and 93% in the external validation data set. Metastatic cancers were identified with a 96% accuracy in the external data set. HiTAIC is a user-friendly web-based application through https://sites.dartmouth.edu/salaslabhitaic/. In conclusion, we developed HiTAIC, a DNA methylation-based algorithm, to trace tumor tissue of origin in primary and metastasized cancers. The high accuracy and resolution of tumor tracing using HiTAIC holds promise for clinical assistance in identifying cancer of unknown origin.

人类癌症的细胞组成和起源部位具有异质性。癌症转移产生了迁移肿瘤细胞起源不明的难题。追踪原发癌和转移癌的起源组织和肿瘤类型对临床意义至关重要。DNA甲基化改变在癌变过程中起着至关重要的作用,标志着细胞命运的分化,因此可以用来追踪肿瘤组织的起源。在这项研究中,我们采用了一种新的肿瘤类型特异性层次模型,利用基因组尺度的DNA甲基化数据开发了一个多层感知器模型HiTAIC,以高分辨率、准确性和特异性追踪7735个肿瘤数据中来自23个组织部位的27种癌症的起源组织和肿瘤类型。在追踪原发性癌症起源时,HiTAIC在测试集的准确度为99%,在外部验证数据集的准确度为93%。在外部数据集中,转移性癌症的识别准确率为96%。HiTAIC是一个用户友好的基于web的应用程序,通过https://sites.dartmouth.edu/salaslabhitaic/。总之,我们开发了一种基于DNA甲基化的算法HiTAIC,用于追踪原发性和转移性癌症的肿瘤组织起源。使用HiTAIC进行肿瘤追踪的高准确性和高分辨率有望为临床鉴定未知来源的癌症提供帮助。
{"title":"<b>HiTAIC: hi</b>erarchical tumor artificial intelligence classifier traces tissue of origin and tumor type in primary and metastasized tumors using DNA methylation.","authors":"Ze Zhang,&nbsp;Yunrui Lu,&nbsp;Soroush Vosoughi,&nbsp;Joshua J Levy,&nbsp;Brock C Christensen,&nbsp;Lucas A Salas","doi":"10.1093/narcan/zcad017","DOIUrl":"https://doi.org/10.1093/narcan/zcad017","url":null,"abstract":"<p><p>Human cancers are heterogenous by their cell composition and origination site. Cancer metastasis generates the conundrum of the unknown origin of migrated tumor cells. Tracing tissue of origin and tumor type in primary and metastasized cancer is vital for clinical significance. DNA methylation alterations play a crucial role in carcinogenesis and mark cell fate differentiation, thus can be used to trace tumor tissue of origin. In this study, we employed a novel tumor-type-specific hierarchical model using genome-scale DNA methylation data to develop a multilayer perceptron model, HiTAIC, to trace tissue of origin and tumor type in 27 cancers from 23 tissue sites in data from 7735 tumors with high resolution, accuracy, and specificity. In tracing primary cancer origin, HiTAIC accuracy was 99% in the test set and 93% in the external validation data set. Metastatic cancers were identified with a 96% accuracy in the external data set. HiTAIC is a user-friendly web-based application through https://sites.dartmouth.edu/salaslabhitaic/. In conclusion, we developed HiTAIC, a DNA methylation-based algorithm, to trace tumor tissue of origin in primary and metastasized cancers. The high accuracy and resolution of tumor tracing using HiTAIC holds promise for clinical assistance in identifying cancer of unknown origin.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/6b/49/zcad017.PMC10113876.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9443818","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: 1
CrypticProteinDB: an integrated database of proteome and immunopeptidome derived non-canonical cancer proteins. CrypticProteinDB:一个蛋白质组和免疫肽来源的非匿名癌症蛋白质的综合数据库。
Pub Date : 2023-06-01 DOI: 10.1093/narcan/zcad024
Ghofran Othoum, Christopher A Maher

Translated non-canonical proteins derived from noncoding regions or alternative open reading frames (ORFs) can contribute to critical and diverse cellular processes. In the context of cancer, they also represent an under-appreciated source of targets for cancer immunotherapy through their tumor-enriched expression or by harboring somatic mutations that produce neoantigens. Here, we introduce the largest integration and proteogenomic analysis of novel peptides to assess the prevalence of non-canonical ORFs (ncORFs) in more than 900 patient proteomes and 26 immunopeptidome datasets across 14 cancer types. The integrative proteogenomic analysis of whole-cell proteomes and immunopeptidomes revealed peptide support for a nonredundant set of 9760 upstream, downstream, and out-of-frame ncORFs in protein coding genes and 12811 in noncoding RNAs. Notably, 6486 ncORFs were derived from differentially expressed genes and 340 were ubiquitously translated across eight or more cancers. The analysis also led to the discovery of thirty-four epitopes and eight neoantigens from non-canonical proteins in two cohorts as novel cancer immunotargets. Collectively, our analysis integrated both bottom-up proteogenomic and targeted peptide validation to illustrate the prevalence of translated non-canonical proteins in cancer and to provide a resource for the prioritization of novel proteins supported by proteomic, immunopeptidomic, genomic and transcriptomic data, available at https://www.maherlab.com/crypticproteindb.

衍生自非编码区或替代开放阅读框(ORF)的翻译非经典蛋白可以促进关键和多样的细胞过程。在癌症的背景下,它们还通过富集肿瘤的表达或携带产生新抗原的体细胞突变,代表了癌症免疫疗法靶点的低估来源。在此,我们介绍了新肽的最大整合和蛋白基因组分析,以评估非匿名ORF(ncORF)在14种癌症类型的900多个患者蛋白质组和26个免疫肽数据集中的患病率。全细胞蛋白质组和免疫肽的综合蛋白基因组分析显示,肽支持蛋白质编码基因中的9760个上游、下游和框架外ncORF和非编码RNA中的12811个非冗余序列。值得注意的是,6486个ncORF来源于差异表达基因,340个在八种或多种癌症中普遍翻译。该分析还发现了34个表位和8个新抗原,它们来自两个队列中的非匿名蛋白,作为新的癌症免疫靶点。总之,我们的分析综合了自下而上的蛋白基因组学和靶向肽验证,以说明翻译的非匿名蛋白在癌症中的流行情况,并为蛋白质组学、免疫肽组学、基因组学和转录组学数据支持的新蛋白的优先顺序提供资源,可在https://www.maherlab.com/crypticproteindb.
{"title":"CrypticProteinDB: an integrated database of proteome and immunopeptidome derived non-canonical cancer proteins.","authors":"Ghofran Othoum, Christopher A Maher","doi":"10.1093/narcan/zcad024","DOIUrl":"10.1093/narcan/zcad024","url":null,"abstract":"<p><p>Translated non-canonical proteins derived from noncoding regions or alternative open reading frames (ORFs) can contribute to critical and diverse cellular processes. In the context of cancer, they also represent an under-appreciated source of targets for cancer immunotherapy through their tumor-enriched expression or by harboring somatic mutations that produce neoantigens. Here, we introduce the largest integration and proteogenomic analysis of novel peptides to assess the prevalence of non-canonical ORFs (ncORFs) in more than 900 patient proteomes and 26 immunopeptidome datasets across 14 cancer types. The integrative proteogenomic analysis of whole-cell proteomes and immunopeptidomes revealed peptide support for a nonredundant set of 9760 upstream, downstream, and out-of-frame ncORFs in protein coding genes and 12811 in noncoding RNAs. Notably, 6486 ncORFs were derived from differentially expressed genes and 340 were ubiquitously translated across eight or more cancers. The analysis also led to the discovery of thirty-four epitopes and eight neoantigens from non-canonical proteins in two cohorts as novel cancer immunotargets. Collectively, our analysis integrated both bottom-up proteogenomic and targeted peptide validation to illustrate the prevalence of translated non-canonical proteins in cancer and to provide a resource for the prioritization of novel proteins supported by proteomic, immunopeptidomic, genomic and transcriptomic data, available at https://www.maherlab.com/crypticproteindb.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10233886/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9584060","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
Over-expression of ADAR1 in mice does not initiate or accelerate cancer formation in vivo. ADAR1在小鼠体内的过度表达不会启动或加速癌症的形成。
Pub Date : 2023-06-01 DOI: 10.1093/narcan/zcad023
Shannon Mendez Ruiz, Alistair M Chalk, Ankita Goradia, Jacki Heraud-Farlow, Carl R Walkley

Adenosine to inosine editing (A-to-I) in regions of double stranded RNA (dsRNA) is mediated by adenosine deaminase acting on RNA 1 (ADAR1) or ADAR2. ADAR1 and A-to-I editing levels are increased in many human cancers. Inhibition of ADAR1 has emerged as a high priority oncology target, however, whether ADAR1 overexpression enables cancer initiation or progression has not been directly tested. We established a series of in vivo models to allow overexpression of full-length ADAR1, or its individual isoforms, to test if increased ADAR1 expression was oncogenic. Widespread over-expression of ADAR1 or the p110 or p150 isoforms individually as sole lesions was well tolerated and did not result in cancer initiation. Therefore, ADAR1 overexpression alone is not sufficient to initiate cancer. We demonstrate that endogenous ADAR1 and A-to-I editing increased upon immortalization in murine cells, consistent with the observations from human cancers. We tested if ADAR1 over-expression could co-operate with cancer initiated by loss of tumour suppressors using a model of osteosarcoma. We did not see a disease potentiating or modifying effect of overexpressing ADAR1 or its isoforms in the models assessed. We conclude that increased ADAR1 expression and A-to-I editing in cancers is most likely a consequence of tumor formation.

双链RNA (dsRNA)区域的腺苷-肌苷编辑(A-to-I)是由作用于RNA 1 (ADAR1)或ADAR2的腺苷脱氨酶介导的。ADAR1和A-to-I编辑水平在许多人类癌症中升高。抑制ADAR1已成为一个高度优先的肿瘤靶点,然而,ADAR1过表达是否会导致癌症的发生或进展尚未直接测试。我们建立了一系列体内模型,允许过表达全长ADAR1或其单个亚型,以测试ADAR1表达增加是否致癌。作为单一病变,ADAR1或p110或p150亚型的广泛过表达具有良好的耐受性,不会导致癌症的发生。因此,仅ADAR1过表达并不足以引发癌症。我们证明内源性ADAR1和A-to-I编辑在小鼠细胞的永生化过程中增加,与人类癌症的观察结果一致。我们使用骨肉瘤模型测试了ADAR1过表达是否与肿瘤抑制因子缺失引发的癌症有关。在评估的模型中,我们没有看到过表达ADAR1或其亚型的疾病增强或修饰作用。我们得出结论,癌症中ADAR1表达和a -to- i编辑的增加很可能是肿瘤形成的结果。
{"title":"Over-expression of ADAR1 in mice does not initiate or accelerate cancer formation <i>in vivo</i>.","authors":"Shannon Mendez Ruiz,&nbsp;Alistair M Chalk,&nbsp;Ankita Goradia,&nbsp;Jacki Heraud-Farlow,&nbsp;Carl R Walkley","doi":"10.1093/narcan/zcad023","DOIUrl":"https://doi.org/10.1093/narcan/zcad023","url":null,"abstract":"<p><p>Adenosine to inosine editing (A-to-I) in regions of double stranded RNA (dsRNA) is mediated by adenosine deaminase acting on RNA 1 (ADAR1) or ADAR2. ADAR1 and A-to-I editing levels are increased in many human cancers. Inhibition of ADAR1 has emerged as a high priority oncology target, however, whether ADAR1 overexpression enables cancer initiation or progression has not been directly tested. We established a series of <i>in vivo</i> models to allow overexpression of full-length ADAR1, or its individual isoforms, to test if increased ADAR1 expression was oncogenic. Widespread over-expression of ADAR1 or the p110 or p150 isoforms individually as sole lesions was well tolerated and did not result in cancer initiation. Therefore, ADAR1 overexpression alone is not sufficient to initiate cancer. We demonstrate that endogenous ADAR1 and A-to-I editing increased upon immortalization in murine cells, consistent with the observations from human cancers. We tested if ADAR1 over-expression could co-operate with cancer initiated by loss of tumour suppressors using a model of osteosarcoma. We did not see a disease potentiating or modifying effect of overexpressing ADAR1 or its isoforms in the models assessed. We conclude that increased ADAR1 expression and A-to-I editing in cancers is most likely a consequence of tumor formation.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10233902/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9584063","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: 5
Alterations of ribosomal RNA pseudouridylation in human breast cancer. 人类乳腺癌中核糖体 RNA 伪苷酸化的变化
Pub Date : 2023-05-30 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad026
Chiara Barozzi, Federico Zacchini, Angelo Gianluca Corradini, Monica Morara, Margherita Serra, Veronica De Sanctis, Roberto Bertorelli, Erik Dassi, Lorenzo Montanaro

RNA modifications are key regulatory factors for several biological and pathological processes. They are abundantly represented on ribosomal RNA (rRNA), where they contribute to regulate ribosomal function in mRNA translation. Altered RNA modification pathways have been linked to tumorigenesis as well as to other human diseases. In this study we quantitatively evaluated the site-specific pseudouridylation pattern in rRNA in breast cancer samples exploiting the RBS-Seq technique involving RNA bisulfite treatment coupled with a new NGS approach. We found a wide variability among patients at different sites. The most dysregulated positions in tumors turned out to be hypermodified with respect to a reference RNA. As for 2'O-methylation level of rRNA modification, we detected variable and stable pseudouridine sites, with the most stable sites being the most evolutionary conserved. We also observed that pseudouridylation levels at specific sites are related to some clinical and bio-pathological tumor features and they are able to distinguish different patient clusters. This study is the first example of the contribution that newly available high-throughput approaches for site specific pseudouridine detection can provide to the understanding of the intrinsic ribosomal changes occurring in human tumors.

RNA 修饰是多种生物和病理过程的关键调节因子。它们大量存在于核糖体 RNA(rRNA)上,在 mRNA 翻译过程中起到调节核糖体功能的作用。RNA 修饰途径的改变与肿瘤发生以及其他人类疾病有关。在这项研究中,我们利用 RBS-Seq 技术(包括 RNA 亚硫酸氢盐处理)和一种新的 NGS 方法,定量评估了乳腺癌样本中 rRNA 的特异位点假酸化模式。我们发现不同部位的患者之间存在很大差异。与参考 RNA 相比,肿瘤中调控最紊乱的位置出现了高修饰。至于 rRNA 修饰的 2'O- 甲基化水平,我们检测到了可变和稳定的假尿苷位点,其中最稳定的位点在进化过程中最为保守。我们还观察到,特定位点的假尿苷化水平与一些临床和生物病理肿瘤特征有关,它们能够区分不同的患者群。这项研究是新近出现的高通量位点特异性假尿嘧啶检测方法为了解人类肿瘤发生的核糖体内在变化所做贡献的第一个实例。
{"title":"Alterations of ribosomal RNA pseudouridylation in human breast cancer.","authors":"Chiara Barozzi, Federico Zacchini, Angelo Gianluca Corradini, Monica Morara, Margherita Serra, Veronica De Sanctis, Roberto Bertorelli, Erik Dassi, Lorenzo Montanaro","doi":"10.1093/narcan/zcad026","DOIUrl":"10.1093/narcan/zcad026","url":null,"abstract":"<p><p>RNA modifications are key regulatory factors for several biological and pathological processes. They are abundantly represented on ribosomal RNA (rRNA), where they contribute to regulate ribosomal function in mRNA translation. Altered RNA modification pathways have been linked to tumorigenesis as well as to other human diseases. In this study we quantitatively evaluated the site-specific pseudouridylation pattern in rRNA in breast cancer samples exploiting the RBS-Seq technique involving RNA bisulfite treatment coupled with a new NGS approach. We found a wide variability among patients at different sites. The most dysregulated positions in tumors turned out to be hypermodified with respect to a reference RNA. As for 2'O-methylation level of rRNA modification, we detected variable and stable pseudouridine sites, with the most stable sites being the most evolutionary conserved. We also observed that pseudouridylation levels at specific sites are related to some clinical and bio-pathological tumor features and they are able to distinguish different patient clusters. This study is the first example of the contribution that newly available high-throughput approaches for site specific pseudouridine detection can provide to the understanding of the intrinsic ribosomal changes occurring in human tumors.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10227372/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9570838","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
Characterization of cell-type specific circular RNAs associated with colorectal cancer metastasis. 与结直肠癌转移相关的细胞类型特异性环状 RNA 的特征。
Pub Date : 2023-05-19 eCollection Date: 2023-06-01 DOI: 10.1093/narcan/zcad021
Sidi Zhao, Amy Ly, Jacqueline L Mudd, Emily B Rozycki, Jace Webster, Emily Coonrod, Ghofran Othoum, Jingqin Luo, Ha X Dang, Ryan C Fields, Christopher A Maher

Colorectal cancer (CRC) is the most common gastrointestinal malignancy and a leading cause of cancer deaths in the United States. More than half of CRC patients develop metastatic disease (mCRC) with an average 5-year survival rate of 13%. Circular RNAs (circRNAs) have recently emerged as important tumorigenesis regulators; however, their role in mCRC progression remains poorly characterized. Further, little is known about their cell-type specificity to elucidate their functions in the tumor microenvironment (TME). To address this, we performed total RNA sequencing (RNA-seq) on 30 matched normal, primary and metastatic samples from 14 mCRC patients. Additionally, five CRC cell lines were sequenced to construct a circRNA catalog in CRC. We detected 47 869 circRNAs, with 51% previously unannotated in CRC and 14% novel candidates when compared to existing circRNA databases. We identified 362 circRNAs differentially expressed in primary and/or metastatic tissues, termed circular RNAs associated with metastasis (CRAMS). We performed cell-type deconvolution using published single-cell RNA-seq datasets and applied a non-negative least squares statistical model to estimate cell-type specific circRNA expression. This predicted 667 circRNAs as exclusively expressed in a single cell type. Collectively, this serves as a valuable resource, TMECircDB (accessible at https://www.maherlab.com/tmecircdb-overview), for functional characterization of circRNAs in mCRC, specifically in the TME.

结肠直肠癌(CRC)是最常见的胃肠道恶性肿瘤,也是美国癌症死亡的主要原因。半数以上的 CRC 患者会出现转移性疾病(mCRC),平均 5 年生存率为 13%。环状 RNAs(circRNAs)最近已成为重要的肿瘤发生调控因子;然而,它们在 mCRC 进展过程中的作用仍鲜为人知。此外,人们对它们的细胞类型特异性知之甚少,无法阐明它们在肿瘤微环境(TME)中的功能。为了解决这个问题,我们对来自 14 名 mCRC 患者的 30 份匹配的正常、原发和转移样本进行了总 RNA 测序(RNA-seq)。此外,我们还对 5 个 CRC 细胞系进行了测序,以构建 CRC 的 circRNA 目录。与现有的 circRNA 数据库相比,我们检测到了 47 869 个 circRNA,其中 51% 以前未在 CRC 中标注过,14% 是新的候选。我们发现了 362 个在原发和/或转移组织中差异表达的 circRNA,称为与转移相关的环状 RNA(CRAMS)。我们利用已发表的单细胞 RNA-seq 数据集进行了细胞类型解卷积,并应用非负最小二乘法统计模型估计了细胞类型特异性 circRNA 的表达。结果预测有 667 种 circRNA 只在一种细胞类型中表达。总而言之,TMECircDB(可在 https://www.maherlab.com/tmecircdb-overview 上访问)是一种宝贵的资源,可用于对 mCRC 中的 circRNA(尤其是 TME 中的 circRNA)进行功能表征。
{"title":"Characterization of cell-type specific circular RNAs associated with colorectal cancer metastasis.","authors":"Sidi Zhao, Amy Ly, Jacqueline L Mudd, Emily B Rozycki, Jace Webster, Emily Coonrod, Ghofran Othoum, Jingqin Luo, Ha X Dang, Ryan C Fields, Christopher A Maher","doi":"10.1093/narcan/zcad021","DOIUrl":"10.1093/narcan/zcad021","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is the most common gastrointestinal malignancy and a leading cause of cancer deaths in the United States. More than half of CRC patients develop metastatic disease (mCRC) with an average 5-year survival rate of 13%. Circular RNAs (circRNAs) have recently emerged as important tumorigenesis regulators; however, their role in mCRC progression remains poorly characterized. Further, little is known about their cell-type specificity to elucidate their functions in the tumor microenvironment (TME). To address this, we performed total RNA sequencing (RNA-seq) on 30 matched normal, primary and metastatic samples from 14 mCRC patients. Additionally, five CRC cell lines were sequenced to construct a circRNA catalog in CRC. We detected 47 869 circRNAs, with 51% previously unannotated in CRC and 14% novel candidates when compared to existing circRNA databases. We identified 362 circRNAs differentially expressed in primary and/or metastatic tissues, termed circular RNAs associated with metastasis (CRAMS). We performed cell-type deconvolution using published single-cell RNA-seq datasets and applied a non-negative least squares statistical model to estimate cell-type specific circRNA expression. This predicted 667 circRNAs as exclusively expressed in a single cell type. Collectively, this serves as a valuable resource, TMECircDB (accessible at https://www.maherlab.com/tmecircdb-overview), for functional characterization of circRNAs in mCRC, specifically in the TME.</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://www.ncbi.nlm.nih.gov/pmc/articles/PMC10198730/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10305723","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