首页 > 最新文献

NAR Cancer最新文献

英文 中文
Correction to 'The DNA repair function of BCL11A suppresses senescence and promotes continued proliferation of triple-negative breast cancer cells'. 修正“BCL11A的DNA修复功能抑制衰老并促进三阴性乳腺癌细胞的持续增殖”。
Pub Date : 2023-03-01 DOI: 10.1093/narcan/zcad008

[This corrects the article DOI: 10.1093/narcan/zcac028.].

[更正文章DOI: 10.1093/narcan/zcac028.]。
{"title":"Correction to 'The DNA repair function of BCL11A suppresses senescence and promotes continued proliferation of triple-negative breast cancer cells'.","authors":"","doi":"10.1093/narcan/zcad008","DOIUrl":"https://doi.org/10.1093/narcan/zcad008","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1093/narcan/zcac028.].</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/8d/ad/zcad008.PMC9900421.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9244036","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
Mono-ADP-ribosylation by PARP10 and PARP14 in genome stability. PARP10和PARP14在基因组稳定性中的单ADP-核糖基化作用
Pub Date : 2023-02-20 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad009
Ashna Dhoonmoon, Claudia M Nicolae

ADP-ribosylation is a post-translational modification involved in a variety of processes including DNA damage repair, transcriptional regulation, and cellular proliferation. Depending on the number of ADP moieties transferred to target proteins, ADP-ribosylation can be classified either as mono-ADP-ribosylation (MARylation) or poly-ADP-ribosylation (PARylation). This post-translational modification is catalyzed by enzymes known as ADP-ribosyltransferases (ARTs), which include the poly (ADP-ribose)-polymerase (PARP) superfamily of proteins. Certain members of the PARP family including PARP1 and PARP2 have been extensively studied and assessed as therapeutic targets. However, the other members of the PARP family of protein are not as well studied but have gained attention in recent years given findings suggesting their roles in an increasing number of cellular processes. Among these other members are PARP10 and PARP14, which have gradually emerged as key players in maintenance of genomic stability and carcinogenesis. PARP10 and PARP14 catalyze the transfer of a single ADP moiety to target proteins. Here, we summarize the current knowledge on MARylation in DNA repair and cancer, focusing on PARP10 and PARP14. We highlight the roles of PARP10 and PARP14 in cancer progression and response to chemotherapeutics and briefly discuss currently known PARP10 and PARP14 inhibitors.

ADP-ribosylation 是一种参与 DNA 损伤修复、转录调控和细胞增殖等多种过程的翻译后修饰。根据转移到目标蛋白质上的 ADP 分子的数量,ADP-核糖基化可分为单 ADP-核糖基化(MARylation)和多 ADP-核糖基化(PARylation)。这种翻译后修饰由称为 ADP-核糖基转移酶(ARTs)的酶催化,其中包括多(ADP-核糖)聚合酶(PARP)超家族蛋白。包括 PARP1 和 PARP2 在内的 PARP 家族的某些成员已被广泛研究并评估为治疗靶点。然而,PARP 家族蛋白质的其他成员虽然研究得不那么深入,但近年来由于研究结果表明它们在越来越多的细胞过程中发挥作用,因而受到了关注。其中,PARP10 和 PARP14 已逐渐成为维持基因组稳定性和致癌的关键角色。PARP10 和 PARP14 催化单个 ADP 分子向靶蛋白的转移。在此,我们总结了目前有关 MARylation 在 DNA 修复和癌症中的作用的知识,重点是 PARP10 和 PARP14。我们强调了 PARP10 和 PARP14 在癌症进展和化疗反应中的作用,并简要讨论了目前已知的 PARP10 和 PARP14 抑制剂。
{"title":"Mono-ADP-ribosylation by PARP10 and PARP14 in genome stability.","authors":"Ashna Dhoonmoon, Claudia M Nicolae","doi":"10.1093/narcan/zcad009","DOIUrl":"10.1093/narcan/zcad009","url":null,"abstract":"<p><p>ADP-ribosylation is a post-translational modification involved in a variety of processes including DNA damage repair, transcriptional regulation, and cellular proliferation. Depending on the number of ADP moieties transferred to target proteins, ADP-ribosylation can be classified either as mono-ADP-ribosylation (MARylation) or poly-ADP-ribosylation (PARylation). This post-translational modification is catalyzed by enzymes known as ADP-ribosyltransferases (ARTs), which include the poly (ADP-ribose)-polymerase (PARP) superfamily of proteins. Certain members of the PARP family including PARP1 and PARP2 have been extensively studied and assessed as therapeutic targets. However, the other members of the PARP family of protein are not as well studied but have gained attention in recent years given findings suggesting their roles in an increasing number of cellular processes. Among these other members are PARP10 and PARP14, which have gradually emerged as key players in maintenance of genomic stability and carcinogenesis. PARP10 and PARP14 catalyze the transfer of a single ADP moiety to target proteins. Here, we summarize the current knowledge on MARylation in DNA repair and cancer, focusing on PARP10 and PARP14. We highlight the roles of PARP10 and PARP14 in cancer progression and response to chemotherapeutics and briefly discuss currently known PARP10 and PARP14 inhibitors.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9940457/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9759076","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
Roles of trans-lesion synthesis (TLS) DNA polymerases in tumorigenesis and cancer therapy. 反式离子合成(TLS)DNA 聚合酶在肿瘤发生和癌症治疗中的作用。
Pub Date : 2023-02-06 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad005
Jay Anand, Lilly Chiou, Carly Sciandra, Xingyuan Zhang, Jiyong Hong, Di Wu, Pei Zhou, Cyrus Vaziri

DNA damage tolerance and mutagenesis are hallmarks and enabling characteristics of neoplastic cells that drive tumorigenesis and allow cancer cells to resist therapy. The 'Y-family' trans-lesion synthesis (TLS) DNA polymerases enable cells to replicate damaged genomes, thereby conferring DNA damage tolerance. Moreover, Y-family DNA polymerases are inherently error-prone and cause mutations. Therefore, TLS DNA polymerases are potential mediators of important tumorigenic phenotypes. The skin cancer-propensity syndrome xeroderma pigmentosum-variant (XPV) results from defects in the Y-family DNA Polymerase Pol eta (Polη) and compensatory deployment of alternative inappropriate DNA polymerases. However, the extent to which dysregulated TLS contributes to the underlying etiology of other human cancers is unclear. Here we consider the broad impact of TLS polymerases on tumorigenesis and cancer therapy. We survey the ways in which TLS DNA polymerases are pathologically altered in cancer. We summarize evidence that TLS polymerases shape cancer genomes, and review studies implicating dysregulated TLS as a driver of carcinogenesis. Because many cancer treatment regimens comprise DNA-damaging agents, pharmacological inhibition of TLS is an attractive strategy for sensitizing tumors to genotoxic therapies. Therefore, we discuss the pharmacological tractability of the TLS pathway and summarize recent progress on development of TLS inhibitors for therapeutic purposes.

DNA 损伤耐受性和突变是肿瘤细胞的标志和有利特征,它们推动肿瘤发生并使癌细胞能够抵抗治疗。Y家族 "反式离子合成(TLS)DNA聚合酶使细胞能够复制受损基因组,从而赋予DNA损伤耐受性。此外,Y-家族 DNA 聚合酶本身容易出错并导致突变。因此,TLS DNA 聚合酶是重要致瘤表型的潜在媒介。皮肤癌倾向综合征色素沉着病变异型(XPV)是由 Y-家族 DNA 聚合酶 Pol eta(Polη)的缺陷和替代性不适当 DNA 聚合酶的补偿性部署造成的。然而,TLS失调在多大程度上导致了其他人类癌症的潜在病因尚不清楚。在此,我们探讨了 TLS 聚合酶对肿瘤发生和癌症治疗的广泛影响。我们调查了 TLS DNA 聚合酶在癌症中发生病理改变的方式。我们总结了 TLS 聚合酶塑造癌症基因组的证据,并回顾了有关 TLS 失调是致癌驱动因素的研究。由于许多癌症治疗方案都包含 DNA 损伤剂,因此对 TLS 进行药理抑制是使肿瘤对基因毒性疗法敏感的一种有吸引力的策略。因此,我们讨论了 TLS 通路的药理学可操作性,并总结了用于治疗目的的 TLS 抑制剂的最新研发进展。
{"title":"Roles of trans-lesion synthesis (TLS) DNA polymerases in tumorigenesis and cancer therapy.","authors":"Jay Anand, Lilly Chiou, Carly Sciandra, Xingyuan Zhang, Jiyong Hong, Di Wu, Pei Zhou, Cyrus Vaziri","doi":"10.1093/narcan/zcad005","DOIUrl":"10.1093/narcan/zcad005","url":null,"abstract":"<p><p>DNA damage tolerance and mutagenesis are hallmarks and enabling characteristics of neoplastic cells that drive tumorigenesis and allow cancer cells to resist therapy. The 'Y-family' trans-lesion synthesis (TLS) DNA polymerases enable cells to replicate damaged genomes, thereby conferring DNA damage tolerance. Moreover, Y-family DNA polymerases are inherently error-prone and cause mutations. Therefore, TLS DNA polymerases are potential mediators of important tumorigenic phenotypes. The skin cancer-propensity syndrome <i>xeroderma pigmentosum-variant</i> (XPV) results from defects in the Y-family DNA Polymerase Pol eta (Polη) and compensatory deployment of alternative inappropriate DNA polymerases. However, the extent to which dysregulated TLS contributes to the underlying etiology of other human cancers is unclear. Here we consider the broad impact of TLS polymerases on tumorigenesis and cancer therapy. We survey the ways in which TLS DNA polymerases are pathologically altered in cancer. We summarize evidence that TLS polymerases shape cancer genomes, and review studies implicating dysregulated TLS as a driver of carcinogenesis. Because many cancer treatment regimens comprise DNA-damaging agents, pharmacological inhibition of TLS is an attractive strategy for sensitizing tumors to genotoxic therapies. Therefore, we discuss the pharmacological tractability of the TLS pathway and summarize recent progress on development of TLS inhibitors for therapeutic purposes.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/6d/65/zcad005.PMC9900426.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10062219","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
APE2: catalytic function and synthetic lethality draw attention as a cancer therapy target. APE2:催化功能和合成致死性引起人们对癌症治疗靶点的关注。
Pub Date : 2023-02-06 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad006
Anne McMahon, Jianjun Zhao, Shan Yan

AP endonuclease 2 (APE2, APEX2 or APN2) is an emerging critical protein involved in genome and epigenome integrity. Whereas its catalytic function as a nuclease in DNA repair is widely accepted, recent studies have elucidated the function and mechanism of APE2 in the immune response and DNA damage response. Several genome-wide screens have identified APE2 as a synthetic lethal target for deficiencies of BRCA1, BRCA2 or TDP1 in cancer cells. Due to its overexpression in several cancer types, APE2 is proposed as an oncogene and could serve as prognostic marker of overall survival of cancer treatment. However, it remains to be discovered whether and how APE2 catalytic function and synthetic lethality can be modulated and manipulated as a cancer therapy target. In this review, we provide a current understanding of alterations and expression of APE2 in cancer, the function of APE2 in the immune response, and mechanisms of APE2 in ATR/Chk1 DNA damage response. We also summarize the role of APE2 in DNA repair pathways in the removal of heterogenous and complexed 3'-termini and MMEJ. Finally, we provide an updated perspective on how APE2 may be targeted for cancer therapy and future directions of APE2 studies in cancer biology.

AP 内切酶 2(APE2、APEX2 或 APN2)是一种参与基因组和表观基因组完整性的新兴关键蛋白。APE2 作为核酸酶在 DNA 修复中的催化功能已被广泛接受,而最近的研究则阐明了 APE2 在免疫反应和 DNA 损伤反应中的功能和机制。一些全基因组筛选发现,APE2 是癌细胞中 BRCA1、BRCA2 或 TDP1 缺陷的合成致死靶点。由于 APE2 在几种癌症类型中的过表达,APE2 被认为是一种癌基因,可作为癌症治疗总生存期的预后标志。然而,APE2 的催化功能和合成致死性是否以及如何作为癌症治疗靶点进行调节和操作仍有待发现。在这篇综述中,我们将介绍目前对 APE2 在癌症中的改变和表达、APE2 在免疫反应中的功能以及 APE2 在 ATR/Chk1 DNA 损伤反应中的机制的理解。我们还总结了 APE2 在 DNA 修复通路中清除异源和复合 3'-termini 以及 MMEJ 的作用。最后,我们从一个最新的角度探讨了如何将 APE2 作为癌症治疗的靶点以及 APE2 在癌症生物学中的未来研究方向。
{"title":"APE2: catalytic function and synthetic lethality draw attention as a cancer therapy target.","authors":"Anne McMahon, Jianjun Zhao, Shan Yan","doi":"10.1093/narcan/zcad006","DOIUrl":"10.1093/narcan/zcad006","url":null,"abstract":"<p><p>AP endonuclease 2 (APE2, APEX2 or APN2) is an emerging critical protein involved in genome and epigenome integrity. Whereas its catalytic function as a nuclease in DNA repair is widely accepted, recent studies have elucidated the function and mechanism of APE2 in the immune response and DNA damage response. Several genome-wide screens have identified APE2 as a synthetic lethal target for deficiencies of BRCA1, BRCA2 or TDP1 in cancer cells. Due to its overexpression in several cancer types, APE2 is proposed as an oncogene and could serve as prognostic marker of overall survival of cancer treatment. However, it remains to be discovered whether and how APE2 catalytic function and synthetic lethality can be modulated and manipulated as a cancer therapy target. In this review, we provide a current understanding of alterations and expression of APE2 in cancer, the function of APE2 in the immune response, and mechanisms of APE2 in ATR/Chk1 DNA damage response. We also summarize the role of APE2 in DNA repair pathways in the removal of heterogenous and complexed 3'-termini and MMEJ. Finally, we provide an updated perspective on how APE2 may be targeted for cancer therapy and future directions of APE2 studies in cancer biology.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9900424/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9759056","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
Tumor-suppressive disruption of cancer subtype-associated super enhancer circuits by small molecule treatment. 通过小分子治疗破坏与癌症亚型相关的超级增强子回路,从而抑制肿瘤。
Pub Date : 2023-02-06 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad007
Anke Koeniger, Pierfrancesco Polo, Anna Brichkina, Florian Finkernagel, Alexander Visekruna, Andrea Nist, Thorsten Stiewe, Michael Daude, Wibke E Diederich, Thomas M Gress, Till Adhikary, Matthias Lauth

Transcriptional cancer subtypes which correlate with traits such as tumor growth, drug sensitivity or the chances of relapse and metastasis, have been described for several malignancies. The core regulatory circuits (CRCs) defining these subtypes are established by chromatin super enhancers (SEs) driving key transcription factors (TFs) specific for the particular cell state. In neuroblastoma (NB), one of the most frequent solid pediatric cancer entities, two major SE-directed molecular subtypes have been described: A more lineage-committed adrenergic (ADRN) and a mesenchymal (MES) subtype. Here, we found that a small isoxazole molecule (ISX), a frequently used pro-neural drug, reprogrammed SE activity and switched NB cells from an ADRN subtype towards a growth-retarded MES-like state. The MES-like state shared strong transcriptional overlap with ganglioneuroma (GN), a benign and highly differentiated tumor of the neural crest. Mechanistically, ISX suppressed chromatin binding of N-MYC, a CRC-amplifying transcription factor, resulting in loss of key ADRN subtype-enriched components such as N-MYC itself, PHOX2B and ALK, while concomitently, MES subtype markers were induced. Globally, ISX treatment installed a chromatin accessibility landscape typically associated with low risk NB. In summary, we provide evidence that CRCs and cancer subtype reprogramming might be amenable to future therapeutic targeting.

一些恶性肿瘤的转录癌症亚型与肿瘤生长、药物敏感性或复发和转移几率等特征相关。定义这些亚型的核心调控回路(CRC)是由驱动特定细胞状态的关键转录因子(TF)的染色质超级增强子(SE)建立的。神经母细胞瘤(NB)是最常见的小儿实体瘤之一,目前已描述了两种主要的SE定向分子亚型:一种是线粒体肾上腺素能亚型(ADRN),另一种是间质亚型(MES)。在这里,我们发现一种常用的促神经药物--异噁唑小分子(ISX)能重编程 SE 的活性,并将 NB 细胞从 ADRN 亚型转换为生长迟缓的 MES 样态。MES样状态与神经节细胞瘤(GN)有很强的转录重叠,后者是神经嵴的一种高分化良性肿瘤。从机理上讲,ISX抑制了N-MYC(一种CRC扩增转录因子)的染色质结合,导致N-MYC本身、PHOX2B和ALK等ADRN亚型丰富的关键成分缺失,同时诱导了MES亚型标志物。从整体上看,ISX 处理设置的染色质可及性景观通常与低风险 NB 相关。总之,我们提供的证据表明,CRC 和癌症亚型重编程可能适合未来的靶向治疗。
{"title":"Tumor-suppressive disruption of cancer subtype-associated super enhancer circuits by small molecule treatment.","authors":"Anke Koeniger, Pierfrancesco Polo, Anna Brichkina, Florian Finkernagel, Alexander Visekruna, Andrea Nist, Thorsten Stiewe, Michael Daude, Wibke E Diederich, Thomas M Gress, Till Adhikary, Matthias Lauth","doi":"10.1093/narcan/zcad007","DOIUrl":"10.1093/narcan/zcad007","url":null,"abstract":"<p><p>Transcriptional cancer subtypes which correlate with traits such as tumor growth, drug sensitivity or the chances of relapse and metastasis, have been described for several malignancies. The core regulatory circuits (CRCs) defining these subtypes are established by chromatin super enhancers (SEs) driving key transcription factors (TFs) specific for the particular cell state. In neuroblastoma (NB), one of the most frequent solid pediatric cancer entities, two major SE-directed molecular subtypes have been described: A more lineage-committed adrenergic (ADRN) and a mesenchymal (MES) subtype. Here, we found that a small isoxazole molecule (ISX), a frequently used pro-neural drug, reprogrammed SE activity and switched NB cells from an ADRN subtype towards a growth-retarded MES-like state. The MES-like state shared strong transcriptional overlap with ganglioneuroma (GN), a benign and highly differentiated tumor of the neural crest. Mechanistically, ISX suppressed chromatin binding of N-MYC, a CRC-amplifying transcription factor, resulting in loss of key ADRN subtype-enriched components such as N-MYC itself, PHOX2B and ALK, while concomitently, MES subtype markers were induced. Globally, ISX treatment installed a chromatin accessibility landscape typically associated with low risk NB. In summary, we provide evidence that CRCs and cancer subtype reprogramming might be amenable to future therapeutic targeting.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9900422/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10684759","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
Ku-DNA binding inhibitors modulate the DNA damage response in response to DNA double-strand breaks. Ku-DNA结合抑制剂调节对DNA双链断裂的DNA损伤反应。
Pub Date : 2023-02-06 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad003
Pamela L Mendoza-Munoz, Navnath S Gavande, Pamela S VanderVere-Carozza, Katherine S Pawelczak, Joseph R Dynlacht, Joy E Garrett, John J Turchi

The DNA-dependent protein kinase (DNA-PK) plays a critical role in the DNA damage response (DDR) and non-homologous end joining (NHEJ) double-strand break (DSB) repair pathways. Consequently, DNA-PK is a validated therapeutic target for cancer treatment in certain DNA repair-deficient cancers and in combination with ionizing radiation (IR). We have previously reported the discovery and development of a novel class of DNA-PK inhibitors with a unique mechanism of action, blocking the Ku 70/80 heterodimer interaction with DNA. These Ku-DNA binding inhibitors (Ku-DBi's) display nanomolar activity in vitro, inhibit cellular DNA-PK, NHEJ-catalyzed DSB repair and sensitize non-small cell lung cancer (NSCLC) cells to DSB-inducing agents. In this study, we demonstrate that chemical inhibition of the Ku-DNA interaction potentiates the cellular effects of bleomycin and IR via p53 phosphorylation through the activation of the ATM pathway. This response is concomitant with a reduction of DNA-PK catalytic subunit (DNA-PKcs) autophosphorylation at S2056 and a time-dependent increase in H2AX phosphorylation at S139. These results are consistent with Ku-DBi's abrogating DNA-PKcs autophosphorylation to impact DSB repair and DDR signaling through a novel mechanism of action, and thus represent a promising anticancer therapeutic strategy in combination with DNA DSB-inducing agents.

DNA依赖性蛋白激酶(DNA-PK)在DNA损伤反应(DDR)和非同源末端连接(NHEJ)双链断裂(DSB)修复途径中起着关键作用。因此,DNA-PK是某些DNA修复缺陷型癌症中癌症治疗的有效治疗靶点,并与电离辐射(IR)相结合。我们之前已经报道了一类新型DNA-PK抑制剂的发现和开发,该抑制剂具有独特的作用机制,阻断Ku 70/80异二聚体与DNA的相互作用。这些Ku-DNA结合抑制剂(Ku-DBi’s)在体外显示纳摩尔活性,抑制细胞DNA-PK、NHEJ-催化的DSB修复,并使非小细胞肺癌癌症(NSCLC)细胞对DSB诱导剂敏感。在这项研究中,我们证明Ku-DNA相互作用的化学抑制通过激活ATM途径,通过p53磷酸化增强博来霉素和IR的细胞效应。这种反应伴随着在S2056处DNA-PK催化亚基(DNA-PKC)自磷酸化的减少和在S139处H2AX磷酸化的时间依赖性增加。这些结果与Ku-DBi通过一种新的作用机制消除DNA PKcs自磷酸化以影响DSB修复和DDR信号传导相一致,因此与DNA DSB诱导剂相结合是一种有前途的抗癌治疗策略。
{"title":"Ku-DNA binding inhibitors modulate the DNA damage response in response to DNA double-strand breaks.","authors":"Pamela L Mendoza-Munoz, Navnath S Gavande, Pamela S VanderVere-Carozza, Katherine S Pawelczak, Joseph R Dynlacht, Joy E Garrett, John J Turchi","doi":"10.1093/narcan/zcad003","DOIUrl":"10.1093/narcan/zcad003","url":null,"abstract":"<p><p>The DNA-dependent protein kinase (DNA-PK) plays a critical role in the DNA damage response (DDR) and non-homologous end joining (NHEJ) double-strand break (DSB) repair pathways. Consequently, DNA-PK is a validated therapeutic target for cancer treatment in certain DNA repair-deficient cancers and in combination with ionizing radiation (IR). We have previously reported the discovery and development of a novel class of DNA-PK inhibitors with a unique mechanism of action, blocking the Ku 70/80 heterodimer interaction with DNA. These Ku-DNA binding inhibitors (Ku-DBi's) display nanomolar activity <i>in vitro</i>, inhibit cellular DNA-PK, NHEJ-catalyzed DSB repair and sensitize non-small cell lung cancer (NSCLC) cells to DSB-inducing agents. In this study, we demonstrate that chemical inhibition of the Ku-DNA interaction potentiates the cellular effects of bleomycin and IR via p53 phosphorylation through the activation of the ATM pathway. This response is concomitant with a reduction of DNA-PK catalytic subunit (DNA-PKcs) autophosphorylation at S2056 and a time-dependent increase in H2AX phosphorylation at S139. These results are consistent with Ku-DBi's abrogating DNA-PKcs autophosphorylation to impact DSB repair and DDR signaling through a novel mechanism of action, and thus represent a promising anticancer therapeutic strategy in combination with DNA DSB-inducing agents.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9900423/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10684758","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
CAFuncAPA: a knowledgebase for systematic functional annotations of APA events in human cancers. CAFuncAPA:人类癌症APA事件的系统功能注释的知识库。
Pub Date : 2023-01-23 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad004
Kexin Huang, Sijia Wu, Xiaotong Yang, Tiangang Wang, Xi Liu, Xiaobo Zhou, Liyu Huang

Alternative polyadenylation (APA) is a widespread posttranscriptional regulation process. APA generates diverse mRNA isoforms with different 3' UTR lengths, affecting mRNA expression, miRNA binding regulation and alternative splicing events. Previous studies have demonstrated the important roles of APA in tumorigenesis and cancer progression through diverse aspects. Thus, a comprehensive functional landscape of diverse APA events would aid in a better understanding of the underlying mechanisms related to APA in human cancers. Here, we built CAFuncAPA (https://relab.xidian.edu.cn/CAFuncAPA/) to systematically annotate the functions of 15478 APA events in human pan-cancers. Specifically, we first identified APA events associated with cancer survival and tumor progression. We annotated the potential downstream effects of APA on genes/isoforms expression, regulation of miRNAs, RNA binding proteins (RBPs) and alternative splicing events. Moreover, we also identified up-regulators of APA events, including the effects of genetic variants on poly(A) sites and RBPs, as well as the effect of methylation phenotypes on APA events. These findings suggested that CAFuncAPA can be a helpful resource for a better understanding of APA regulators and potential functions in cancer biology.

选择性多腺苷酸化(APA)是一个广泛的转录后调控过程。APA产生具有不同3’UTR长度的不同mRNA亚型,影响mRNA表达、miRNA结合调节和选择性剪接事件。先前的研究已经通过不同方面证明了APA在肿瘤发生和癌症进展中的重要作用。因此,对不同APA事件的全面功能景观将有助于更好地理解人类癌症中与APA相关的潜在机制。在这里,我们建造了CAFuncAPA(https://relab.xidian.edu.cn/CAFuncAPA/)以系统地注释15478个APA事件在人类泛癌中的功能。具体而言,我们首先确定了与癌症生存和肿瘤进展相关的APA事件。我们注释了APA对基因/异构体表达、miRNA、RNA结合蛋白(RBPs)和选择性剪接事件的潜在下游影响。此外,我们还确定了APA事件的上调因子,包括遗传变异对poly(A)位点和RBPs的影响,以及甲基化表型对APA事件影响。这些发现表明,CAFuncAPA可以成为更好地了解APA调节因子和癌症生物学中潜在功能的有用资源。
{"title":"CAFuncAPA: a knowledgebase for systematic functional annotations of APA events in human cancers.","authors":"Kexin Huang, Sijia Wu, Xiaotong Yang, Tiangang Wang, Xi Liu, Xiaobo Zhou, Liyu Huang","doi":"10.1093/narcan/zcad004","DOIUrl":"10.1093/narcan/zcad004","url":null,"abstract":"<p><p>Alternative polyadenylation (APA) is a widespread posttranscriptional regulation process. APA generates diverse mRNA isoforms with different 3' UTR lengths, affecting mRNA expression, miRNA binding regulation and alternative splicing events. Previous studies have demonstrated the important roles of APA in tumorigenesis and cancer progression through diverse aspects. Thus, a comprehensive functional landscape of diverse APA events would aid in a better understanding of the underlying mechanisms related to APA in human cancers. Here, we built CAFuncAPA (https://relab.xidian.edu.cn/CAFuncAPA/) to systematically annotate the functions of 15478 APA events in human pan-cancers. Specifically, we first identified APA events associated with cancer survival and tumor progression. We annotated the potential downstream effects of APA on genes/isoforms expression, regulation of miRNAs, RNA binding proteins (RBPs) and alternative splicing events. Moreover, we also identified up-regulators of APA events, including the effects of genetic variants on poly(A) sites and RBPs, as well as the effect of methylation phenotypes on APA events. These findings suggested that CAFuncAPA can be a helpful resource for a better understanding of APA regulators and potential functions in cancer biology.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9869079/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10671892","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
Oncogenic ERRB2 signals through the AP-1 transcription factor to control mesenchymal-like properties of oesophageal adenocarcinoma. 致癌物质ERRB2通过AP-1转录因子发出信号,控制食管腺癌的间质样特性。
Pub Date : 2023-01-23 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad001
Samuel Ogden, Ibrahim Ahmed, Shen-Hsi Yang, Paul Fullwood, Chiara Francavilla, Andrew D Sharrocks

Oesophageal adenocarcinoma (OAC) is a deadly disease with poor survival statistics and few targeted therapies available. One of the most common molecular aberrations in OAC is amplification or activation of the gene encoding the receptor tyrosine kinase ERBB2, and ERBB2 is targeted in the clinic for this subset of patients. However, the downstream consequences of these ERBB2 activating events are not well understood. Here we used a combination of phosphoproteomics, open chromatin profiling and transcriptome analysis on cell line models and patient-derived datasets to interrogate the molecular pathways operating downstream from ERBB2. Integrated analysis of these data sets converge on a model where dysregulated ERBB2 signalling is mediated at the transcriptional level by the transcription factor AP-1. AP-1 in turn controls cell behaviour by acting on cohorts of genes that regulate cell migration and adhesion, features often associated with EMT. Our study therefore provides a valuable resource for the cancer cell signalling community and reveals novel molecular determinants underlying the dysregulated behaviour of OAC cells.

食管腺癌(OAC)是一种致命疾病,存活率很低,而且几乎没有靶向疗法。食管腺癌最常见的分子畸变之一是编码受体酪氨酸激酶ERBB2的基因扩增或激活,ERBB2在临床上是这部分患者的靶向药物。然而,人们对这些ERBB2激活事件的下游后果还不甚了解。在这里,我们在细胞系模型和患者数据集上结合使用了磷蛋白组学、开放染色质图谱分析和转录组分析,以探究ERBB2下游的分子通路。对这些数据集的综合分析汇聚到一个模型上,即ERBB2信号失调是由转录因子AP-1在转录水平上介导的。AP-1反过来又通过作用于调控细胞迁移和粘附的基因群来控制细胞行为,这些基因群通常与EMT相关。因此,我们的研究为癌细胞信号社区提供了宝贵的资源,并揭示了 OAC 细胞行为失调的新分子决定因素。
{"title":"Oncogenic ERRB2 signals through the AP-1 transcription factor to control mesenchymal-like properties of oesophageal adenocarcinoma.","authors":"Samuel Ogden, Ibrahim Ahmed, Shen-Hsi Yang, Paul Fullwood, Chiara Francavilla, Andrew D Sharrocks","doi":"10.1093/narcan/zcad001","DOIUrl":"10.1093/narcan/zcad001","url":null,"abstract":"<p><p>Oesophageal adenocarcinoma (OAC) is a deadly disease with poor survival statistics and few targeted therapies available. One of the most common molecular aberrations in OAC is amplification or activation of the gene encoding the receptor tyrosine kinase ERBB2, and ERBB2 is targeted in the clinic for this subset of patients. However, the downstream consequences of these ERBB2 activating events are not well understood. Here we used a combination of phosphoproteomics, open chromatin profiling and transcriptome analysis on cell line models and patient-derived datasets to interrogate the molecular pathways operating downstream from ERBB2. Integrated analysis of these data sets converge on a model where dysregulated ERBB2 signalling is mediated at the transcriptional level by the transcription factor AP-1. AP-1 in turn controls cell behaviour by acting on cohorts of genes that regulate cell migration and adhesion, features often associated with EMT. Our study therefore provides a valuable resource for the cancer cell signalling community and reveals novel molecular determinants underlying the dysregulated behaviour of OAC cells.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9869078/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10255331","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
EEPD1 promotes repair of oxidatively-stressed replication forks. EEPD1促进氧化应激复制叉的修复。
Pub Date : 2023-01-18 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcac044
Aruna S Jaiswal, Hyun-Suk Kim, Orlando D Schärer, Neelam Sharma, Elizabeth A Williamson, Gayathri Srinivasan, Linda Phillips, Kimi Kong, Shailee Arya, Anurag Misra, Arijit Dutta, Yogesh Gupta, Christi A Walter, Sandeep Burma, Satya Narayan, Patrick Sung, Jac A Nickoloff, Robert Hromas

Unrepaired oxidatively-stressed replication forks can lead to chromosomal instability and neoplastic transformation or cell death. To meet these challenges cells have evolved a robust mechanism to repair oxidative genomic DNA damage through the base excision repair (BER) pathway, but less is known about repair of oxidative damage at replication forks. We found that depletion or genetic deletion of EEPD1 decreases clonogenic cell survival after oxidative DNA damage. We demonstrate that EEPD1 is recruited to replication forks stressed by oxidative damage induced by H2O2 and that EEPD1 promotes replication fork repair and restart and decreases chromosomal abnormalities after such damage. EEPD1 binds to abasic DNA structures and promotes resolution of genomic abasic sites after oxidative stress. We further observed that restoration of expression of EEPD1 via expression vector transfection restores cell survival and suppresses chromosomal abnormalities induced by oxidative stress in EEPD1-depleted cells. Consistent with this, we found that EEPD1 preserves replication fork integrity by preventing oxidatively-stressed unrepaired fork fusion, thereby decreasing chromosome instability and mitotic abnormalities. Our results indicate a novel role for EEPD1 in replication fork preservation and maintenance of chromosomal stability during oxidative stress.

未配对的氧化应激复制叉可导致染色体不稳定和肿瘤转化或细胞死亡。为了应对这些挑战,细胞已经进化出一种强大的机制,通过碱基切除修复(BER)途径修复基因组DNA的氧化损伤,但对复制叉处氧化损伤的修复知之甚少。我们发现,EEPD1的缺失或基因缺失会降低DNA氧化损伤后的克隆细胞存活率。我们证明,EEPD1被招募到由H2O2诱导的氧化损伤所应激的复制叉中,并且EEPD1促进复制叉的修复和重启,并减少这种损伤后的染色体异常。EEPD1与碱性DNA结构结合,并促进氧化应激后基因组碱性位点的分解。我们进一步观察到,在EEPD1缺失的细胞中,通过表达载体转染恢复EEPD1的表达可恢复细胞存活并抑制氧化应激诱导的染色体异常。与此一致,我们发现EEPD1通过防止氧化应激的未修复叉融合来保持复制叉的完整性,从而减少染色体不稳定性和有丝分裂异常。我们的研究结果表明,在氧化应激期间,EEPD1在复制叉保存和维持染色体稳定性方面具有新的作用。
{"title":"EEPD1 promotes repair of oxidatively-stressed replication forks.","authors":"Aruna S Jaiswal, Hyun-Suk Kim, Orlando D Schärer, Neelam Sharma, Elizabeth A Williamson, Gayathri Srinivasan, Linda Phillips, Kimi Kong, Shailee Arya, Anurag Misra, Arijit Dutta, Yogesh Gupta, Christi A Walter, Sandeep Burma, Satya Narayan, Patrick Sung, Jac A Nickoloff, Robert Hromas","doi":"10.1093/narcan/zcac044","DOIUrl":"10.1093/narcan/zcac044","url":null,"abstract":"<p><p>Unrepaired oxidatively-stressed replication forks can lead to chromosomal instability and neoplastic transformation or cell death. To meet these challenges cells have evolved a robust mechanism to repair oxidative genomic DNA damage through the base excision repair (BER) pathway, but less is known about repair of oxidative damage at replication forks. We found that depletion or genetic deletion of EEPD1 decreases clonogenic cell survival after oxidative DNA damage. We demonstrate that EEPD1 is recruited to replication forks stressed by oxidative damage induced by H<sub>2</sub>O<sub>2</sub> and that EEPD1 promotes replication fork repair and restart and decreases chromosomal abnormalities after such damage. EEPD1 binds to abasic DNA structures and promotes resolution of genomic abasic sites after oxidative stress. We further observed that restoration of expression of EEPD1 via expression vector transfection restores cell survival and suppresses chromosomal abnormalities induced by oxidative stress in EEPD1-depleted cells. Consistent with this, we found that EEPD1 preserves replication fork integrity by preventing oxidatively-stressed unrepaired fork fusion, thereby decreasing chromosome instability and mitotic abnormalities. Our results indicate a novel role for EEPD1 in replication fork preservation and maintenance of chromosomal stability during oxidative stress.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9846428/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9771122","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
Longitudinal evaluation of serum microRNAs as biomarkers for neuroblastoma burden and therapeutic p53 reactivation. 将血清微RNA作为神经母细胞瘤负担和治疗性p53再激活的生物标记物的纵向评估。
Pub Date : 2023-01-18 eCollection Date: 2023-03-01 DOI: 10.1093/narcan/zcad002
Alan Van Goethem, Jill Deleu, Nurten Yigit, Celine Everaert, Myrthala Moreno-Smith, Sanjeev A Vasudevan, Fjoralba Zeka, Fleur Demuynck, Eveline Barbieri, Frank Speleman, Pieter Mestdagh, Jason Shohet, Jo Vandesompele, Tom Van Maerken

Accurate assessment of treatment response and residual disease is indispensable for the evaluation of cancer treatment efficacy. However, performing tissue biopsies for longitudinal follow-up poses a major challenge in the management of solid tumours like neuroblastoma. In the present study, we evaluated whether circulating miRNAs are suitable to monitor neuroblastoma tumour burden and whether treatment-induced changes of miRNA abundance in the tumour are detectable in serum. We performed small RNA sequencing on longitudinally collected serum samples from mice carrying orthotopic neuroblastoma xenografts that were exposed to treatment with idasanutlin or temsirolimus. We identified 57 serum miRNAs to be differentially expressed upon xenograft tumour manifestation, out of which 21 were also found specifically expressed in the serum of human high-risk neuroblastoma patients. The murine serum levels of these 57 miRNAs correlated with tumour tissue expression and tumour volume, suggesting potential utility for monitoring tumour burden. In addition, we describe serum miRNAs that dynamically respond to p53 activation following treatment of engrafted mice with idasanutlin. We identified idasanutlin-induced serum miRNA expression changes upon one day and 11 days of treatment. By limiting to miRNAs with a tumour-related induction, we put forward hsa-miR-34a-5p as a potential pharmacodynamic biomarker of p53 activation in serum.

准确评估治疗反应和残留疾病对于评价癌症治疗效果不可或缺。然而,在神经母细胞瘤等实体瘤的治疗中,进行组织活检以进行纵向随访是一项重大挑战。在本研究中,我们评估了循环 miRNA 是否适用于监测神经母细胞瘤的肿瘤负荷,以及是否能在血清中检测到治疗引起的肿瘤中 miRNA 丰度的变化。我们对纵向收集的血清样本进行了小RNA测序,这些样本来自接受伊达司林或替西莫司治疗的携带正位神经母细胞瘤异种移植物的小鼠。我们发现 57 种血清 miRNA 在异种移植肿瘤显现时有不同表达,其中 21 种在人类高危神经母细胞瘤患者的血清中也有特异表达。小鼠血清中这 57 种 miRNA 的水平与肿瘤组织表达和肿瘤体积相关,这表明它们在监测肿瘤负荷方面具有潜在的作用。此外,我们还描述了用idasanutlin处理移植小鼠后,血清miRNA对p53激活的动态响应。我们确定了idasanutlin诱导的血清miRNA在治疗1天和11天后的表达变化。通过限制与肿瘤相关的miRNA,我们提出了hsa-miR-34a-5p作为血清中p53激活的潜在药效学生物标记物。
{"title":"Longitudinal evaluation of serum microRNAs as biomarkers for neuroblastoma burden and therapeutic p53 reactivation.","authors":"Alan Van Goethem, Jill Deleu, Nurten Yigit, Celine Everaert, Myrthala Moreno-Smith, Sanjeev A Vasudevan, Fjoralba Zeka, Fleur Demuynck, Eveline Barbieri, Frank Speleman, Pieter Mestdagh, Jason Shohet, Jo Vandesompele, Tom Van Maerken","doi":"10.1093/narcan/zcad002","DOIUrl":"10.1093/narcan/zcad002","url":null,"abstract":"<p><p>Accurate assessment of treatment response and residual disease is indispensable for the evaluation of cancer treatment efficacy. However, performing tissue biopsies for longitudinal follow-up poses a major challenge in the management of solid tumours like neuroblastoma. In the present study, we evaluated whether circulating miRNAs are suitable to monitor neuroblastoma tumour burden and whether treatment-induced changes of miRNA abundance in the tumour are detectable in serum. We performed small RNA sequencing on longitudinally collected serum samples from mice carrying orthotopic neuroblastoma xenografts that were exposed to treatment with idasanutlin or temsirolimus. We identified 57 serum miRNAs to be differentially expressed upon xenograft tumour manifestation, out of which 21 were also found specifically expressed in the serum of human high-risk neuroblastoma patients. The murine serum levels of these 57 miRNAs correlated with tumour tissue expression and tumour volume, suggesting potential utility for monitoring tumour burden. In addition, we describe serum miRNAs that dynamically respond to p53 activation following treatment of engrafted mice with idasanutlin. We identified idasanutlin-induced serum miRNA expression changes upon one day and 11 days of treatment. By limiting to miRNAs with a tumour-related induction, we put forward hsa-miR-34a-5p as a potential pharmacodynamic biomarker of p53 activation in serum.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/12/21/zcad002.PMC9846426.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10635063","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