首页 > 最新文献

Cancer Cell International最新文献

英文 中文
The role and application of small extracellular vesicles in glioma. 细胞外小泡在胶质瘤中的作用和应用。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-29 DOI: 10.1186/s12935-024-03389-z
Zhihao Yang, HaoYuan Wu, ZhiWei Wang, ErBao Bian, Bing Zhao

Small extracellular vesicles (sEVs) are cell-derived, nanometer-sized particles enclosed by a lipid bilayer. All kinds of biological molecules, including proteins, DNA fragments, RNA, lipids, and metabolites, can be selectively loaded into sEVs and transmitted to recipient cells that are near and distant. Growing shreds of evidence show the significant biological function and the clinical significance of sEVs in cancers. Numerous recent studies have validated that sEVs play an important role in tumor progression and can be utilized to diagnose, stage, grading, and monitor early tumors. In addition, sEVs have also served as drug delivery nanocarriers and cancer vaccines. Although it is still infancy, the field of basic and translational research based on sEVs has grown rapidly. In this review, we summarize the latest research on sEVs in gliomas, including their role in the malignant biological function of gliomas, and the potential of sEVs in non-invasive diagnostic and therapeutic approaches, i.e., as nanocarriers for drug or gene delivery and cancer vaccines.

细胞外小泡(sEVs)是一种源自细胞、由脂质双分子层包裹的纳米级颗粒。各种生物分子,包括蛋白质、DNA 片段、RNA、脂质和代谢物,都可以选择性地装载到 sEVs 中,并传输到远近的受体细胞。越来越多的证据表明,sEVs 在癌症中具有重要的生物学功能和临床意义。最近的大量研究证实,sEVs 在肿瘤进展过程中发挥着重要作用,可用于诊断、分期、分级和监测早期肿瘤。此外,sEVs 还可用作给药纳米载体和癌症疫苗。基于 sEVs 的基础研究和转化研究虽然仍处于起步阶段,但发展迅速。在这篇综述中,我们总结了有关神经胶质瘤中 sEVs 的最新研究,包括它们在神经胶质瘤恶性生物功能中的作用,以及 sEVs 在非侵入性诊断和治疗方法中的潜力,即作为药物或基因递送的纳米载体和癌症疫苗。
{"title":"The role and application of small extracellular vesicles in glioma.","authors":"Zhihao Yang, HaoYuan Wu, ZhiWei Wang, ErBao Bian, Bing Zhao","doi":"10.1186/s12935-024-03389-z","DOIUrl":"10.1186/s12935-024-03389-z","url":null,"abstract":"<p><p>Small extracellular vesicles (sEVs) are cell-derived, nanometer-sized particles enclosed by a lipid bilayer. All kinds of biological molecules, including proteins, DNA fragments, RNA, lipids, and metabolites, can be selectively loaded into sEVs and transmitted to recipient cells that are near and distant. Growing shreds of evidence show the significant biological function and the clinical significance of sEVs in cancers. Numerous recent studies have validated that sEVs play an important role in tumor progression and can be utilized to diagnose, stage, grading, and monitor early tumors. In addition, sEVs have also served as drug delivery nanocarriers and cancer vaccines. Although it is still infancy, the field of basic and translational research based on sEVs has grown rapidly. In this review, we summarize the latest research on sEVs in gliomas, including their role in the malignant biological function of gliomas, and the potential of sEVs in non-invasive diagnostic and therapeutic approaches, i.e., as nanocarriers for drug or gene delivery and cancer vaccines.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11218314/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476004","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting of 3D oral cancer spheroids by αVβ6 integrin using near-infrared peptide-conjugated IRDye 680. 利用近红外肽结合 IRDye 680 靶向αVβ6 整合素的三维口腔癌球体。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-29 DOI: 10.1186/s12935-024-03417-y
L Dirheimer, T Pons, A François, L Lamy, S Cortese, F Marchal, L Bezdetnaya

Background: In the treatment of oral cavity cancer, margin status is one of the most critical prognostic factors. Positive margins are associated with higher local recurrence and lower survival rates. Therefore, the universal goal of oral surgical oncology is to achieve microscopically clear margins. Near-infrared fluorescence guided surgery (FGS) could improve surgical resection using fluorescent probes. αVβ6 integrin has shown great potential for cancer targeting due to its overexpression in oral cancers. Red fluorescent contrast agent IRDye 680 coupled with anti-αVβ6 peptide (IRDye-A20) represents an asset to improve FGS of oral cancer. This study investigates the potential of IRDye-A20 as a selective imaging agent in 3D three-dimensional tongue cancer cells.

Methods: αVβ6 integrin expression was evaluated by RT-qPCR and Western Blotting in 2D HSC-3 human tongue cancer cells and MRC-5 human fibroblasts. Targeting ability of IRDye-A20 was studied in both cell lines by flow cytometry technique. 3D tumor spheroid models, homotypic (HSC-3) and stroma-enriched heterotypic (HSC-3/MRC-5) spheroids were produced by liquid overlay procedure and further characterized using (immuno)histological and fluorescence-based techniques. IRDye-A20 selectivity was evaluated in each type of spheroids and each cell population.

Results: αVβ6 integrin was overexpressed in 2D HSC-3 cancer cells but not in MRC-5 fibroblasts and consistently, only HSC-3 were labelled with IRDye-A20. Round shaped spheroids with an average diameter of 400 μm were produced with a final ratio of 55%/45% between HSC-3 and MRC-5 cells, respectively. Immunofluorescence experiments demonstrated an uniform expression of αVβ6 integrin in homotypic spheroid, while its expression was restricted to cancer cells only in heterotypic spheroid. In stroma-enriched 3D model, Cytokeratin 19 and E-cadherin were expressed only by cancer cells while vimentin and fibronectin were expressed by fibroblasts. Using flow cytometry, we demonstrated that IRDye-A20 labeled the whole homotypic spheroid, while in the heterotypic model all cancer cells were highly fluorescent, with a negligible fluorescence in fibroblasts.

Conclusions: The present study demonstrated an efficient selective targeting of A20FMDV2-conjugated IRDye 680 in 3D tongue cancer cells stroma-enriched spheroids. Thus, IRDye-A20 could be a promising candidate for the future development of the fluorescence-guided surgery of oral cancers.

背景:在口腔癌的治疗中,边缘状态是最关键的预后因素之一。阳性边缘与较高的局部复发率和较低的生存率有关。因此,口腔肿瘤外科的普遍目标是实现显微镜下清晰的边缘。由于αVβ6整合素在口腔癌中的过度表达,它在癌症靶向方面显示出巨大的潜力。红色荧光造影剂 IRDye 680 与抗αVβ6 肽(IRDye-A20)结合使用,可改善口腔癌的 FGS。本研究探讨了 IRDye-A20 在三维立体舌癌细胞中作为选择性成像剂的潜力。流式细胞术研究了 IRDye-A20 在这两种细胞系中的靶向能力。通过液体覆盖法制作了三维肿瘤球体模型、同型球体(HSC-3)和富含基质的异型球体(HSC-3/MRC-5),并使用(免疫)组织学和荧光技术对其进行了进一步表征。结果:αVβ6整合素在二维HSC-3癌细胞中过表达,而在MRC-5成纤维细胞中没有过表达,因此只有HSC-3细胞被IRDye-A20标记。HSC-3和MRC-5细胞的最终比例分别为55%和45%,形成了平均直径为400微米的圆形球体。免疫荧光实验表明,αVβ6整合素在同型球体内均匀表达,而在异型球体内仅局限于癌细胞表达。在基质丰富的三维模型中,只有癌细胞表达细胞角蛋白19和E-cadherin,而成纤维细胞表达波形蛋白和纤连蛋白。利用流式细胞术,我们证明 IRDye-A20 标记了整个同型球体,而在异型模型中,所有癌细胞都发出高荧光,成纤维细胞的荧光可忽略不计:本研究证明了 A20FMDV2 共轭 IRDye 680 在三维舌癌细胞基质丰富球体内的高效选择性靶向作用。因此,IRDye-A20有望成为未来口腔癌荧光引导手术的候选药物。
{"title":"Targeting of 3D oral cancer spheroids by αVβ6 integrin using near-infrared peptide-conjugated IRDye 680.","authors":"L Dirheimer, T Pons, A François, L Lamy, S Cortese, F Marchal, L Bezdetnaya","doi":"10.1186/s12935-024-03417-y","DOIUrl":"10.1186/s12935-024-03417-y","url":null,"abstract":"<p><strong>Background: </strong>In the treatment of oral cavity cancer, margin status is one of the most critical prognostic factors. Positive margins are associated with higher local recurrence and lower survival rates. Therefore, the universal goal of oral surgical oncology is to achieve microscopically clear margins. Near-infrared fluorescence guided surgery (FGS) could improve surgical resection using fluorescent probes. αVβ6 integrin has shown great potential for cancer targeting due to its overexpression in oral cancers. Red fluorescent contrast agent IRDye 680 coupled with anti-αVβ6 peptide (IRDye-A20) represents an asset to improve FGS of oral cancer. This study investigates the potential of IRDye-A20 as a selective imaging agent in 3D three-dimensional tongue cancer cells.</p><p><strong>Methods: </strong>αVβ6 integrin expression was evaluated by RT-qPCR and Western Blotting in 2D HSC-3 human tongue cancer cells and MRC-5 human fibroblasts. Targeting ability of IRDye-A20 was studied in both cell lines by flow cytometry technique. 3D tumor spheroid models, homotypic (HSC-3) and stroma-enriched heterotypic (HSC-3/MRC-5) spheroids were produced by liquid overlay procedure and further characterized using (immuno)histological and fluorescence-based techniques. IRDye-A20 selectivity was evaluated in each type of spheroids and each cell population.</p><p><strong>Results: </strong>αVβ6 integrin was overexpressed in 2D HSC-3 cancer cells but not in MRC-5 fibroblasts and consistently, only HSC-3 were labelled with IRDye-A20. Round shaped spheroids with an average diameter of 400 μm were produced with a final ratio of 55%/45% between HSC-3 and MRC-5 cells, respectively. Immunofluorescence experiments demonstrated an uniform expression of αVβ6 integrin in homotypic spheroid, while its expression was restricted to cancer cells only in heterotypic spheroid. In stroma-enriched 3D model, Cytokeratin 19 and E-cadherin were expressed only by cancer cells while vimentin and fibronectin were expressed by fibroblasts. Using flow cytometry, we demonstrated that IRDye-A20 labeled the whole homotypic spheroid, while in the heterotypic model all cancer cells were highly fluorescent, with a negligible fluorescence in fibroblasts.</p><p><strong>Conclusions: </strong>The present study demonstrated an efficient selective targeting of A20FMDV2-conjugated IRDye 680 in 3D tongue cancer cells stroma-enriched spheroids. Thus, IRDye-A20 could be a promising candidate for the future development of the fluorescence-guided surgery of oral cancers.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11218202/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476003","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pitavastatin sensitizes the EGFR-TKI associated resistance in lung cancer by inhibiting YAP/AKT/BAD-BCL-2 pathway. 匹伐他汀通过抑制YAP/AKT/BAD-BCL-2通路,使肺癌中与表皮生长因子受体-TKI相关的耐药性变得敏感。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-28 DOI: 10.1186/s12935-024-03416-z
Jie Liu, Jialei Fu, Ping Fu, Menghan Liu, Zining Liu, Bao Song

Background: Despite effective strategies, resistance in EGFR mutated lung cancer remains a challenge. Metabolic reprogramming is one of the main mechanisms of tumor drug resistance. A class of drugs known as "statins" inhibit lipid cholesterol metabolism and are widely used in patients with cardiovascular diseases. Previous studies have also documented its ability to improve the therapeutic impact in lung cancer patients who receive EGFR-TKI therapy. Therefore, the effect of statins on targeted drug resistance to lung cancer remains to be investigated.

Methods: Prolonged exposure to gefitinib resulted in the emergence of a resistant lung cancer cell line (PC9GR) from the parental sensitive cell line (PC9), which exhibited a traditional EGFR mutation. The CCK-8 assay was employed to assess the impact of various concentrations of pitavastatin on cellular proliferation. RNA sequencing was conducted to detect differentially expressed genes and their correlated pathways. For the detection of protein expression, Western blot was performed. The antitumor activity of pitavastatin was evaluated in vivo via a xenograft mouse model.

Results: PC9 gefitinib resistant strains were induced by low-dose maintenance. Cell culture and animal-related studies validated that the application of pitavastatin inhibited the proliferation of lung cancer cells, promoted cell apoptosis, and restrained the acquired resistance to EGFR-TKIs. KEGG pathway analysis showed that the hippo/YAP signaling pathway was activated in PC9GR cells relative to PC9 cells, and the YAP expression was inhibited by pitavastatin administration. With YAP RNA interference, pAKT, pBAD and BCL-2 expression was decreased, while BAX expression as increased. Accordingly, YAP down-regulated significantly increased apoptosis and decreased the survival rate of gefitinib-resistant lung cancer cells. After pAKT was increased by SC79, apoptosis of YAP down-regulated cells induced by gefitinib was decreased, and the cell survival rate was increased. Mechanistically, these effects of pitavastatin are associated with the YAP pathway, thereby inhibiting the downstream AKT/BAD-BCL-2 signaling pathway.

Conclusion: Our study provides a molecular basis for the clinical application of the lipid-lowering drug pitavastatin enhances the susceptibility of lung cancer to EGFR-TKI drugs and alleviates drug resistance.

背景:尽管采取了有效的策略,但表皮生长因子受体突变肺癌的耐药性仍是一项挑战。代谢重编程是肿瘤耐药的主要机制之一。一类被称为 "他汀类 "的药物可抑制脂质胆固醇代谢,被广泛用于心血管疾病患者。以往的研究也证明,他汀类药物能够改善接受表皮生长因子受体-TKI治疗的肺癌患者的治疗效果。因此,他汀类药物对肺癌靶向药物耐药性的影响仍有待研究:方法:长期暴露于吉非替尼导致耐药肺癌细胞系(PC9GR)从亲代敏感细胞系(PC9)中产生,后者表现出传统的表皮生长因子受体(EGFR)突变。采用CCK-8测定法评估了不同浓度的匹伐他汀对细胞增殖的影响。通过 RNA 测序检测差异表达基因及其相关通路。为了检测蛋白质的表达,进行了 Western 印迹。通过异种移植小鼠模型评估匹伐他汀的体内抗肿瘤活性:结果:通过低剂量维持诱导出PC9吉非替尼耐药株。细胞培养和动物相关研究证实,应用匹伐他汀可抑制肺癌细胞增殖,促进细胞凋亡,抑制对表皮生长因子受体抑制剂(EGFR-TKIs)的获得性耐药。KEGG通路分析表明,相对于PC9细胞,PC9GR细胞中的hippo/YAP信号通路被激活,服用匹伐他汀后YAP的表达受到抑制。通过对YAP进行RNA干扰,pAKT、pBAD和BCL-2的表达量减少,而BAX的表达量增加。因此,下调 YAP 能显著增加吉非替尼耐药肺癌细胞的凋亡,降低其存活率。通过 SC79 增加 pAKT 后,吉非替尼诱导的 YAP 下调细胞的凋亡减少,细胞存活率增加。从机理上讲,匹伐他汀的这些作用与YAP通路有关,从而抑制了下游的AKT/BAD-BCL-2信号通路:我们的研究为临床应用降脂药物匹伐他汀增强肺癌对EGFR-TKI药物的易感性和缓解耐药性提供了分子基础。
{"title":"Pitavastatin sensitizes the EGFR-TKI associated resistance in lung cancer by inhibiting YAP/AKT/BAD-BCL-2 pathway.","authors":"Jie Liu, Jialei Fu, Ping Fu, Menghan Liu, Zining Liu, Bao Song","doi":"10.1186/s12935-024-03416-z","DOIUrl":"https://doi.org/10.1186/s12935-024-03416-z","url":null,"abstract":"<p><strong>Background: </strong>Despite effective strategies, resistance in EGFR mutated lung cancer remains a challenge. Metabolic reprogramming is one of the main mechanisms of tumor drug resistance. A class of drugs known as \"statins\" inhibit lipid cholesterol metabolism and are widely used in patients with cardiovascular diseases. Previous studies have also documented its ability to improve the therapeutic impact in lung cancer patients who receive EGFR-TKI therapy. Therefore, the effect of statins on targeted drug resistance to lung cancer remains to be investigated.</p><p><strong>Methods: </strong>Prolonged exposure to gefitinib resulted in the emergence of a resistant lung cancer cell line (PC9GR) from the parental sensitive cell line (PC9), which exhibited a traditional EGFR mutation. The CCK-8 assay was employed to assess the impact of various concentrations of pitavastatin on cellular proliferation. RNA sequencing was conducted to detect differentially expressed genes and their correlated pathways. For the detection of protein expression, Western blot was performed. The antitumor activity of pitavastatin was evaluated in vivo via a xenograft mouse model.</p><p><strong>Results: </strong>PC9 gefitinib resistant strains were induced by low-dose maintenance. Cell culture and animal-related studies validated that the application of pitavastatin inhibited the proliferation of lung cancer cells, promoted cell apoptosis, and restrained the acquired resistance to EGFR-TKIs. KEGG pathway analysis showed that the hippo/YAP signaling pathway was activated in PC9GR cells relative to PC9 cells, and the YAP expression was inhibited by pitavastatin administration. With YAP RNA interference, pAKT, pBAD and BCL-2 expression was decreased, while BAX expression as increased. Accordingly, YAP down-regulated significantly increased apoptosis and decreased the survival rate of gefitinib-resistant lung cancer cells. After pAKT was increased by SC79, apoptosis of YAP down-regulated cells induced by gefitinib was decreased, and the cell survival rate was increased. Mechanistically, these effects of pitavastatin are associated with the YAP pathway, thereby inhibiting the downstream AKT/BAD-BCL-2 signaling pathway.</p><p><strong>Conclusion: </strong>Our study provides a molecular basis for the clinical application of the lipid-lowering drug pitavastatin enhances the susceptibility of lung cancer to EGFR-TKI drugs and alleviates drug resistance.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11214206/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466328","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NFIC mediates m6A mRNA methylation to orchestrate transcriptional and post-transcriptional regulation to represses malignant phenotype of non-small cell lung cancer cells. NFIC 介导 m6A mRNA 甲基化,协调转录和转录后调控,抑制非小细胞肺癌细胞的恶性表型。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-28 DOI: 10.1186/s12935-024-03414-1
Kesong Shi, Yani Chen, Ruihua Liu, Xinyao Fu, Hua Guo, Tian Gao, Shu Wang, Le Dou, Jiemin Wang, Yuan Wu, Jiale Yu, Haiquan Yu

Background: Multiple genetic and epigenetic regulatory mechanisms are crucial in the development and tumorigenesis process. Transcriptional regulation often involves intricate relationships and networks with post-transcriptional regulatory molecules, impacting the spatial and temporal expression of genes. However, the synergistic relationship between transcription factors and N6-methyladenosine (m6A) modification in regulating gene expression, as well as their influence on the mechanisms underlying the occurrence and progression of non-small cell lung cancer (NSCLC), requires further investigation. The present study aimed to investigate the synergistic relationship between transcription factors and m6A modification on NSCLC.

Methods: The transcription factor NFIC and its potential genes was screened by analyzing publicly available datasets (ATAC-seq, DNase-seq, and RNA-seq). The association of NFIC and its potential target genes were validated through ChIP-qPCR and dual-luciferase reporter assays. Additionally, the roles of NFIC and its potential genes in NSCLC were detected in vitro and in vivo through silencing and overexpression assays.

Results: Based on multi-omics data, the transcription factor NFIC was identified as a potential tumor suppressor of NSCLC. NFIC was significantly downregulated in both NSCLC tissues and cells, and when NFIC was overexpressed, the malignant phenotype and total m6A content of NSCLC cells was suppressed, while the PI3K/AKT pathway was inactivated. Additionally, we discovered that NFIC inhibits the expression of METTL3 by directly binding to its promoter region, and METTL3 regulates the expression of KAT2A, a histone acetyltransferase, by methylating the m6A site in the 3'UTR of KAT2A mRNA in NSCLC cells. Intriguingly, NFIC was also found to negatively regulate the expression of KAT2A by directly binding to its promoter region.

Conclusions: Our findings demonstrated that NFIC suppresses the malignant phenotype of NSCLC cells by regulating gene expression at both the transcriptional and post-transcriptional levels. A deeper comprehension of the genetic and epigenetic regulatory mechanisms in tumorigenesis would be beneficial for the development of personalized treatment strategies.

背景:多种遗传和表观遗传调控机制在发育和肿瘤发生过程中至关重要。转录调控往往涉及与转录后调控分子之间错综复杂的关系和网络,影响基因在空间和时间上的表达。然而,转录因子与 N6-甲基腺苷(m6A)修饰在调控基因表达方面的协同作用关系,以及它们对非小细胞肺癌(NSCLC)发生和发展机制的影响,还需要进一步研究。本研究旨在探讨转录因子与 m6A 修饰对 NSCLC 的协同作用:方法:通过分析公开数据集(ATAC-seq、DNase-seq和RNA-seq)筛选转录因子NFIC及其潜在基因。通过 ChIP-qPCR 和双荧光素酶报告实验验证了 NFIC 及其潜在靶基因之间的关联。此外,还通过沉默和过表达实验检测了NFIC及其潜在基因在体外和体内NSCLC中的作用:结果:基于多组学数据,转录因子NFIC被鉴定为NSCLC的潜在肿瘤抑制因子。NFIC在NSCLC组织和细胞中均明显下调,当NFIC过表达时,NSCLC细胞的恶性表型和总m6A含量被抑制,同时PI3K/AKT通路失活。此外,我们还发现NFIC通过直接结合METTL3的启动子区域来抑制其表达,而METTL3则通过甲基化KAT2A mRNA的3'UTR中的m6A位点来调控NSCLC细胞中组蛋白乙酰转移酶KAT2A的表达。耐人寻味的是,NFIC还通过直接与KAT2A的启动子区域结合,负向调节KAT2A的表达:我们的研究结果表明,NFIC通过在转录和转录后水平调控基因表达来抑制NSCLC细胞的恶性表型。深入了解肿瘤发生过程中的遗传学和表观遗传学调控机制将有助于制定个性化的治疗策略。
{"title":"NFIC mediates m6A mRNA methylation to orchestrate transcriptional and post-transcriptional regulation to represses malignant phenotype of non-small cell lung cancer cells.","authors":"Kesong Shi, Yani Chen, Ruihua Liu, Xinyao Fu, Hua Guo, Tian Gao, Shu Wang, Le Dou, Jiemin Wang, Yuan Wu, Jiale Yu, Haiquan Yu","doi":"10.1186/s12935-024-03414-1","DOIUrl":"https://doi.org/10.1186/s12935-024-03414-1","url":null,"abstract":"<p><strong>Background: </strong>Multiple genetic and epigenetic regulatory mechanisms are crucial in the development and tumorigenesis process. Transcriptional regulation often involves intricate relationships and networks with post-transcriptional regulatory molecules, impacting the spatial and temporal expression of genes. However, the synergistic relationship between transcription factors and N6-methyladenosine (m6A) modification in regulating gene expression, as well as their influence on the mechanisms underlying the occurrence and progression of non-small cell lung cancer (NSCLC), requires further investigation. The present study aimed to investigate the synergistic relationship between transcription factors and m6A modification on NSCLC.</p><p><strong>Methods: </strong>The transcription factor NFIC and its potential genes was screened by analyzing publicly available datasets (ATAC-seq, DNase-seq, and RNA-seq). The association of NFIC and its potential target genes were validated through ChIP-qPCR and dual-luciferase reporter assays. Additionally, the roles of NFIC and its potential genes in NSCLC were detected in vitro and in vivo through silencing and overexpression assays.</p><p><strong>Results: </strong>Based on multi-omics data, the transcription factor NFIC was identified as a potential tumor suppressor of NSCLC. NFIC was significantly downregulated in both NSCLC tissues and cells, and when NFIC was overexpressed, the malignant phenotype and total m6A content of NSCLC cells was suppressed, while the PI3K/AKT pathway was inactivated. Additionally, we discovered that NFIC inhibits the expression of METTL3 by directly binding to its promoter region, and METTL3 regulates the expression of KAT2A, a histone acetyltransferase, by methylating the m6A site in the 3'UTR of KAT2A mRNA in NSCLC cells. Intriguingly, NFIC was also found to negatively regulate the expression of KAT2A by directly binding to its promoter region.</p><p><strong>Conclusions: </strong>Our findings demonstrated that NFIC suppresses the malignant phenotype of NSCLC cells by regulating gene expression at both the transcriptional and post-transcriptional levels. A deeper comprehension of the genetic and epigenetic regulatory mechanisms in tumorigenesis would be beneficial for the development of personalized treatment strategies.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11212411/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466327","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
WZ-3146 acts as a novel small molecule inhibitor of KIF4A to inhibit glioma progression by inducing apoptosis. WZ-3146 是一种新型的 KIF4A 小分子抑制剂,可通过诱导细胞凋亡抑制胶质瘤的发展。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-27 DOI: 10.1186/s12935-024-03409-y
Tao Yan, Qing Jiang, Guangpu Ni, Haofeng Ma, Yun Meng, Guiqiong Kang, Meifang Xu, Fei Peng, Huadong Li, Xin Chen, Mingguang Wang

Background: Glioma is considered the most common primary malignant tumor of the central nervous system. Although traditional treatments have not achieved satisfactory outcomes, recently, targeted therapies for glioma have shown promising efficacy. However, due to the single-target nature of targeted therapy, traditional targeted therapies are ineffective; thus, novel therapeutic targets are urgently needed.

Methods: The gene expression data for glioma patients were derived from the GEO (GSE4290, GSE50161), TCGA and CGGA databases. Next, the upregulated genes obtained from the above databases were cross-analyzed, finally, 10 overlapping genes (BIRC5, FOXM1, EZH2, CDK1, KIF11, KIF4A, NDC80, PBK, RRM2, and TOP2A) were ultimately screened and only KIF4A expression has the strongest correlation with clinical characteristics in glioma patients. Futher, the TCGA and CGGA database were utilized to explore the correlation of KIF4A expression with glioma prognosis. Then, qRT-PCR and Western blot was used to detect the KIF4A mRNA and protein expression level in glioma cells, respectively. And WZ-3146, the small molecule inhibitor targeting KIF4A, were screened by Cmap analysis. Subsequently, the effect of KIF4A knockdown or WZ-3146 treatment on glioma was measured by the MTT, EdU, Colony formation assay and Transwell assay. Ultimately, GSEA enrichment analysis was performed to find that the apoptotic pathway could be regulated by KIF4A in glioma, in addition, the effect of WZ-3146 on glioma apoptosis was detected by flow cytometry and Western blot.

Results: In the present study, we confirmed that KIF4A is abnormally overexpressed in glioma. In addition, KIF4A overexpression is a key indicator of glioma prognosis; moreover, suppressing KIF4A expression can inhibit glioma progression. We also discovered that WZ-3146, a small molecule inhibitor of KIF4A, can induce apoptosis in glioma cells and exhibit antiglioma effects.

Conclusion: In conclusion, these observations demonstrated that targeting KIF4A can inhibit glioma progression. With further research, WZ-3146, a small molecule inhibitor of KIF4A, could be combined with other molecular targeted drugs to cooperatively inhibit glioma progression.

背景:胶质瘤是中枢神经系统最常见的原发性恶性肿瘤:胶质瘤被认为是中枢神经系统最常见的原发性恶性肿瘤。虽然传统治疗方法的疗效并不令人满意,但最近,胶质瘤靶向疗法显示出了良好的疗效。然而,由于靶向治疗的单靶点性,传统靶向治疗效果不佳,因此迫切需要新的治疗靶点:胶质瘤患者的基因表达数据来自 GEO(GSE4290、GSE50161)、TCGA 和 CGGA 数据库。然后,对上述数据库中的上调基因进行交叉分析,最终筛选出 10 个重叠基因(BIRC5、FOXM1、EZH2、CDK1、KIF11、KIF4A、NDC80、PBK、RRM2 和 TOP2A),其中只有 KIF4A 的表达与胶质瘤患者的临床特征相关性最强。随后,研究人员利用TCGA和CGGA数据库探讨了KIF4A表达与胶质瘤预后的相关性。然后,利用qRT-PCR和Western blot分别检测胶质瘤细胞中KIF4A mRNA和蛋白的表达水平。并通过Cmap分析筛选出靶向KIF4A的小分子抑制剂WZ-3146。随后,通过MTT、EdU、集落形成试验和Transwell试验测定了KIF4A敲除或WZ-3146处理对胶质瘤的影响。最后,通过GSEA富集分析发现胶质瘤中的凋亡通路可能受KIF4A调控,此外,还通过流式细胞术和Western blot检测了WZ-3146对胶质瘤凋亡的影响:结果:本研究证实,KIF4A在胶质瘤中异常过表达。此外,抑制 KIF4A 的表达可抑制胶质瘤的进展。我们还发现,KIF4A 的小分子抑制剂 WZ-3146 可诱导胶质瘤细胞凋亡,并具有抗胶质瘤作用:总之,这些观察结果表明,靶向 KIF4A 可以抑制胶质瘤的发展。随着研究的深入,KIF4A 小分子抑制剂 WZ-3146 可与其他分子靶向药物联合使用,共同抑制胶质瘤的发展。
{"title":"WZ-3146 acts as a novel small molecule inhibitor of KIF4A to inhibit glioma progression by inducing apoptosis.","authors":"Tao Yan, Qing Jiang, Guangpu Ni, Haofeng Ma, Yun Meng, Guiqiong Kang, Meifang Xu, Fei Peng, Huadong Li, Xin Chen, Mingguang Wang","doi":"10.1186/s12935-024-03409-y","DOIUrl":"https://doi.org/10.1186/s12935-024-03409-y","url":null,"abstract":"<p><strong>Background: </strong>Glioma is considered the most common primary malignant tumor of the central nervous system. Although traditional treatments have not achieved satisfactory outcomes, recently, targeted therapies for glioma have shown promising efficacy. However, due to the single-target nature of targeted therapy, traditional targeted therapies are ineffective; thus, novel therapeutic targets are urgently needed.</p><p><strong>Methods: </strong>The gene expression data for glioma patients were derived from the GEO (GSE4290, GSE50161), TCGA and CGGA databases. Next, the upregulated genes obtained from the above databases were cross-analyzed, finally, 10 overlapping genes (BIRC5, FOXM1, EZH2, CDK1, KIF11, KIF4A, NDC80, PBK, RRM2, and TOP2A) were ultimately screened and only KIF4A expression has the strongest correlation with clinical characteristics in glioma patients. Futher, the TCGA and CGGA database were utilized to explore the correlation of KIF4A expression with glioma prognosis. Then, qRT-PCR and Western blot was used to detect the KIF4A mRNA and protein expression level in glioma cells, respectively. And WZ-3146, the small molecule inhibitor targeting KIF4A, were screened by Cmap analysis. Subsequently, the effect of KIF4A knockdown or WZ-3146 treatment on glioma was measured by the MTT, EdU, Colony formation assay and Transwell assay. Ultimately, GSEA enrichment analysis was performed to find that the apoptotic pathway could be regulated by KIF4A in glioma, in addition, the effect of WZ-3146 on glioma apoptosis was detected by flow cytometry and Western blot.</p><p><strong>Results: </strong>In the present study, we confirmed that KIF4A is abnormally overexpressed in glioma. In addition, KIF4A overexpression is a key indicator of glioma prognosis; moreover, suppressing KIF4A expression can inhibit glioma progression. We also discovered that WZ-3146, a small molecule inhibitor of KIF4A, can induce apoptosis in glioma cells and exhibit antiglioma effects.</p><p><strong>Conclusion: </strong>In conclusion, these observations demonstrated that targeting KIF4A can inhibit glioma progression. With further research, WZ-3146, a small molecule inhibitor of KIF4A, could be combined with other molecular targeted drugs to cooperatively inhibit glioma progression.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11209999/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466329","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Breast cancer patient-derived organoids for the investigation of patient-specific tumour evolution. 用于研究患者特异性肿瘤演变的乳腺癌患者衍生器官组织。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-27 DOI: 10.1186/s12935-024-03375-5
Serena Mazzucchelli, Lorena Signati, Letizia Messa, Alma Franceschini, Arianna Bonizzi, Lorenzo Castagnoli, Patrizia Gasparini, Clarissa Consolandi, Eleonora Mangano, Paride Pelucchi, Ingrid Cifola, Tania Camboni, Marco Severgnini, Laura Villani, Barbara Tagliaferri, Stephana Carelli, Serenella M Pupa, Cristina Cereda, Fabio Corsi

Background: A reliable preclinical model of patient-derived organoids (PDOs) was developed in a case study of a 69-year-old woman diagnosed with breast cancer (BC) to investigate the tumour evolution before and after neoadjuvant chemotherapy and surgery. The results were achieved due to the development of PDOs from tissues collected before (O-PRE) and after (O-POST) treatment.

Methods: PDO cultures were characterized by histology, immunohistochemistry (IHC), transmission electron microscopy (TEM), scanning electron microscopy (SEM), confocal microscopy, flow cytometry, real-time PCR, bulk RNA-seq, single-cell RNA sequencing (scRNA-seq) and drug screening.

Results: Both PDO cultures recapitulated the histological and molecular profiles of the original tissues, and they showed typical mammary gland organization, confirming their reliability as a personalized in vitro model. Compared with O-PRE, O-POST had a greater proliferation rate with a significant increase in the Ki67 proliferation index. Moreover O-POST exhibited a more stem-like and aggressive phenotype, with increases in the CD24low/CD44low and EPCAMlow/CD49fhigh cell populations characterized by increased tumour initiation potential and multipotency and metastatic potential in invasive lobular carcinoma. Analysis of ErbB receptor expression indicated a decrease in HER-2 expression coupled with an increase in EGFR expression in O-POST. In this context, deregulation of the PI3K/Akt signalling pathway was assessed by transcriptomic analysis, confirming the altered transcriptional profile. Finally, transcriptomic single-cell analysis identified 11 cell type clusters, highlighting the selection of the luminal component and the decrease in the number of Epithelial-mesenchymal transition cell types in O-POST.

Conclusion: Neoadjuvant treatment contributed to the enrichment of cell populations with luminal phenotypes that were more resistant to chemotherapy in O-POST. PDOs represent an excellent 3D cell model for assessing disease evolution.

背景:通过对一名被诊断为乳腺癌(BC)的69岁妇女的病例研究,建立了一个可靠的临床前患者来源器官组织(PDOs)模型,以研究新辅助化疗和手术前后的肿瘤演变情况。这些结果是通过从治疗前(O-PRE)和治疗后(O-POST)采集的组织中培养 PDOs 得出的:方法:通过组织学、免疫组化(IHC)、透射电子显微镜(TEM)、扫描电子显微镜(SEM)、共聚焦显微镜、流式细胞术、实时 PCR、大量 RNA-seq、单细胞 RNA 测序(scRNA-seq)和药物筛选对 PDO 培养物进行鉴定:结果:两种PDO培养物都再现了原始组织的组织学和分子特征,并表现出典型的乳腺组织,证实了其作为个性化体外模型的可靠性。与O-PRE相比,O-POST的增殖率更高,Ki67增殖指数显著增加。此外,O-POST 表现出更多的干细胞样和侵袭性表型,CD24-低/CD44-低和 EPCAM-低/CD49f-高细胞群增加,其特点是在浸润性小叶癌中肿瘤启动潜能、多能性和转移潜能增加。对 ErbB 受体表达的分析表明,在 O-POST 中,HER-2 表达减少,而 EGFR 表达增加。在这种情况下,通过转录组分析评估了 PI3K/Akt 信号通路的失调,证实了转录谱的改变。最后,转录组单细胞分析确定了11个细胞类型群,突出显示了O-POST中腔内成分的选择和上皮-间质转化细胞类型数量的减少:结论:新辅助治疗促使具有管腔表型的细胞群在O-POST中富集,这些细胞群对化疗更具抵抗力。PDO是评估疾病演变的绝佳三维细胞模型。
{"title":"Breast cancer patient-derived organoids for the investigation of patient-specific tumour evolution.","authors":"Serena Mazzucchelli, Lorena Signati, Letizia Messa, Alma Franceschini, Arianna Bonizzi, Lorenzo Castagnoli, Patrizia Gasparini, Clarissa Consolandi, Eleonora Mangano, Paride Pelucchi, Ingrid Cifola, Tania Camboni, Marco Severgnini, Laura Villani, Barbara Tagliaferri, Stephana Carelli, Serenella M Pupa, Cristina Cereda, Fabio Corsi","doi":"10.1186/s12935-024-03375-5","DOIUrl":"10.1186/s12935-024-03375-5","url":null,"abstract":"<p><strong>Background: </strong>A reliable preclinical model of patient-derived organoids (PDOs) was developed in a case study of a 69-year-old woman diagnosed with breast cancer (BC) to investigate the tumour evolution before and after neoadjuvant chemotherapy and surgery. The results were achieved due to the development of PDOs from tissues collected before (O-PRE) and after (O-POST) treatment.</p><p><strong>Methods: </strong>PDO cultures were characterized by histology, immunohistochemistry (IHC), transmission electron microscopy (TEM), scanning electron microscopy (SEM), confocal microscopy, flow cytometry, real-time PCR, bulk RNA-seq, single-cell RNA sequencing (scRNA-seq) and drug screening.</p><p><strong>Results: </strong>Both PDO cultures recapitulated the histological and molecular profiles of the original tissues, and they showed typical mammary gland organization, confirming their reliability as a personalized in vitro model. Compared with O-PRE, O-POST had a greater proliferation rate with a significant increase in the Ki67 proliferation index. Moreover O-POST exhibited a more stem-like and aggressive phenotype, with increases in the CD24<sup>low</sup>/CD44<sup>low</sup> and EPCAM<sup>low</sup>/CD49f<sup>high</sup> cell populations characterized by increased tumour initiation potential and multipotency and metastatic potential in invasive lobular carcinoma. Analysis of ErbB receptor expression indicated a decrease in HER-2 expression coupled with an increase in EGFR expression in O-POST. In this context, deregulation of the PI3K/Akt signalling pathway was assessed by transcriptomic analysis, confirming the altered transcriptional profile. Finally, transcriptomic single-cell analysis identified 11 cell type clusters, highlighting the selection of the luminal component and the decrease in the number of Epithelial-mesenchymal transition cell types in O-POST.</p><p><strong>Conclusion: </strong>Neoadjuvant treatment contributed to the enrichment of cell populations with luminal phenotypes that were more resistant to chemotherapy in O-POST. PDOs represent an excellent 3D cell model for assessing disease evolution.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11210105/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141455461","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inhibition of ABI2 ubiquitination-dependent degradation suppresses TNBC cell growth via down-regulating PI3K/Akt signaling pathway. 抑制ABI2泛素依赖性降解可通过下调PI3K/Akt信号通路抑制TNBC细胞的生长。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-27 DOI: 10.1186/s12935-024-03407-0
Linlin Lv, Shujing Li, Jie Kang, Yulin Li, Nannan Zhao, Dongman Ye, Fengying Qin, Jing Sun, Tao Yu, Huijian Wu

Triple negative breast cancer (TNBC) is a type of cancer that lacks receptor expression and has complex molecular mechanisms. Recent evidence shows that the ubiquitin-protease system is closely related to TNBC. In this study, we obtain a key ubiquitination regulatory substrate-ABI2 protein by bioinformatics methods, which is also closely related to the survival and prognosis of TNBC. Further, through a series of experiments, we demonstrated that ABI2 expressed at a low level in TNBC tumors, and it has the ability to control cell cycle and inhibit TNBC cell migration, invasion and proliferation. Molecular mechanism studies proved E3 ligase CBLC could increase the ubiquitination degradation of ABI2 protein. Meanwhile, RNA-seq and IP experiments indicated that ABI2, acting as a crucial factor of tumor suppression, can significantly inhibit PI3K/Akt signaling pathway via the interaction with Rho GTPase RAC1. Finally, based on TNBC drug target ABI2, we screened and found that FDA-approved drug Colistimethate sodium(CS) has significant potential in suppressing the proliferation of TNBC cells and inducing cell apoptosis, making it a promising candidate for impeding the progression of TNBC.

三阴性乳腺癌(TNBC)是一种缺乏受体表达且分子机制复杂的癌症。最新证据表明,泛素蛋白酶系统与 TNBC 密切相关。在本研究中,我们通过生物信息学方法获得了一个关键泛素化调控底物--ABI2蛋白,该蛋白也与TNBC的生存和预后密切相关。此外,通过一系列实验证明,ABI2在TNBC肿瘤中低水平表达,具有控制细胞周期、抑制TNBC细胞迁移、侵袭和增殖的作用。分子机制研究证明,E3连接酶CBLC可增加ABI2蛋白的泛素化降解。同时,RNA-seq和IP实验表明,ABI2通过与Rho GTPase RAC1相互作用,可显著抑制PI3K/Akt信号通路,是抑制肿瘤的关键因子。最后,基于TNBC药物靶点ABI2,我们筛选并发现美国FDA批准的药物考来烯胺酸钠(Colistimethate sodium,CS)在抑制TNBC细胞增殖和诱导细胞凋亡方面具有显著潜力,有望成为阻碍TNBC进展的候选药物。
{"title":"Inhibition of ABI2 ubiquitination-dependent degradation suppresses TNBC cell growth via down-regulating PI3K/Akt signaling pathway.","authors":"Linlin Lv, Shujing Li, Jie Kang, Yulin Li, Nannan Zhao, Dongman Ye, Fengying Qin, Jing Sun, Tao Yu, Huijian Wu","doi":"10.1186/s12935-024-03407-0","DOIUrl":"https://doi.org/10.1186/s12935-024-03407-0","url":null,"abstract":"<p><p>Triple negative breast cancer (TNBC) is a type of cancer that lacks receptor expression and has complex molecular mechanisms. Recent evidence shows that the ubiquitin-protease system is closely related to TNBC. In this study, we obtain a key ubiquitination regulatory substrate-ABI2 protein by bioinformatics methods, which is also closely related to the survival and prognosis of TNBC. Further, through a series of experiments, we demonstrated that ABI2 expressed at a low level in TNBC tumors, and it has the ability to control cell cycle and inhibit TNBC cell migration, invasion and proliferation. Molecular mechanism studies proved E3 ligase CBLC could increase the ubiquitination degradation of ABI2 protein. Meanwhile, RNA-seq and IP experiments indicated that ABI2, acting as a crucial factor of tumor suppression, can significantly inhibit PI3K/Akt signaling pathway via the interaction with Rho GTPase RAC1. Finally, based on TNBC drug target ABI2, we screened and found that FDA-approved drug Colistimethate sodium(CS) has significant potential in suppressing the proliferation of TNBC cells and inducing cell apoptosis, making it a promising candidate for impeding the progression of TNBC.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11212232/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141466356","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Antiproliferative effect of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues on IL-6 mediated STAT3 and role of the apoptotic pathway in albino Wistar rats of ethyl carbamate-induced lung carcinoma: In-silico, In-vitro, and In-vivo study. 茚并[1,2-d]噻唑并[3,2-a]嘧啶类似物对 IL-6 介导的 STAT3 的抗增殖作用以及氨基甲酸乙酯诱导的白化 Wistar 大鼠肺癌细胞凋亡途径的作用:硅内、体外和体内研究。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-26 DOI: 10.1186/s12935-024-03390-6
Archana Bharti Sonkar, Abhishek Verma, Sneha Yadav, Rohit Kumar, Jyoti Singh, Amit K Keshari, Soniya Rani, Anurag Kumar, Dharmendra Kumar, Neeraj Kumar Shrivastava, Shubham Rastogi, Mariam K Alamoudi, Mohd Nazam Ansari, Abdulaziz S Saeedan, Gaurav Kaithwas, Sudipta Saha

Lung cancer (LC) ranks second most prevalent cancer in females after breast cancer and second in males after prostate cancer. Based on the GLOBOCAN 2020 report, India represented 5.9% of LC cases and 8.1% of deaths caused by the disease. Several clinical studies have shown that LC occurs because of biological and morphological abnormalities and the involvement of altered level of antioxidants, cytokines, and apoptotic markers. In the present study, we explored the antiproliferative activity of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues against LC using in-vitro, in-silico, and in-vivo models. In-vitro screening against A549 cells revealed compounds 9B (8-methoxy-5-(3,4,5-trimethoxyphenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine) and 12B (5-(4-chlorophenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine) as potential pyrimidine analogues against LC. Compounds 9B and 12B were docked with different molecular targets IL-6, Cyt-C, Caspase9, and Caspase3 using AutoDock Vina 4.1 to evaluate the binding affinity. Subsequently, in-vivo studies were conducted in albino Wistar rats through ethyl-carbamate (EC)- induced LC. 9B and 12B imparted significant effects on physiological (weight variation), and biochemical (anti-oxidant [TBAR's, SOD, ProC, and GSH), lipid (TC, TG, LDL, VLDL, and HDL)], and cytokine (IL-2, IL-6, IL-10, and IL-1β) markers in EC-induced LC in albino Wistar rats. Morphological examination (SEM and H&E) and western blotting (IL-6, STAT3, Cyt-C, BAX, Bcl-2, Caspase3, and caspase9) showed that compounds 9B and 12B had antiproliferative effects. Accordingly, from the in-vitro, in-silico, and in-vivo experimental findings, we concluded that 9B and 12B have significant antiproliferative potential and are potential candidates for further evaluation to meet the requirements of investigation of new drug application.

肺癌(LC)在女性癌症发病率中仅次于乳腺癌,在男性癌症发病率中仅次于前列腺癌。根据 GLOBOCAN 2020 报告,印度占肺癌病例的 5.9%,占肺癌死亡人数的 8.1%。多项临床研究表明,乳腺癌的发生是由于生物学和形态学异常,以及抗氧化剂、细胞因子和细胞凋亡标志物水平的改变。在本研究中,我们采用体外、硅内和体内模型,探讨了茚并[1,2-d]噻唑并[3,2-a]嘧啶类似物对 LC 的抗增殖活性。针对 A549 细胞的体外筛选显示,化合物 9B (8-甲氧基-5-(3,4,5-三甲氧基苯基)-5,6-二氢茚并[1,2-d]噻唑并[3,2-a]嘧啶)和 12B (5-(4-氯苯基)-5,6-二氢茚并[1,2-d]噻唑并[3,2-a]嘧啶)是抗 LC 的潜在嘧啶类似物。使用 AutoDock Vina 4.1 将化合物 9B 和 12B 与不同的分子靶标 IL-6、Cyt-C、Caspase9 和 Caspase3 进行对接,以评估其结合亲和力。随后,在白化 Wistar 大鼠体内通过氨基甲酸乙酯(EC)诱导 LC 进行了体内研究。9B 和 12B 对白化 Wistar 大鼠在氨基甲酸乙酯诱导的 LC 中的生理指标(体重变化)、生化指标(抗氧化剂 [TBAR、SOD、ProC 和 GSH]、血脂指标(TC、TG、LDL、VLDL 和 HDL)]和细胞因子指标(IL-2、IL-6、IL-10 和 IL-1β)均有显著影响。形态学检查(SEM和H&E)和Western印迹(IL-6、STAT3、Cyt-C、BAX、Bcl-2、Caspase3和caspase9)表明,化合物9B和12B具有抗增殖作用。因此,从体外、硅内和体内实验结果来看,我们认为 9B 和 12B 具有显著的抗增殖潜力,是进一步评估的潜在候选化合物,以满足新药申请研究的要求。
{"title":"Antiproliferative effect of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues on IL-6 mediated STAT3 and role of the apoptotic pathway in albino Wistar rats of ethyl carbamate-induced lung carcinoma: In-silico, In-vitro, and In-vivo study.","authors":"Archana Bharti Sonkar, Abhishek Verma, Sneha Yadav, Rohit Kumar, Jyoti Singh, Amit K Keshari, Soniya Rani, Anurag Kumar, Dharmendra Kumar, Neeraj Kumar Shrivastava, Shubham Rastogi, Mariam K Alamoudi, Mohd Nazam Ansari, Abdulaziz S Saeedan, Gaurav Kaithwas, Sudipta Saha","doi":"10.1186/s12935-024-03390-6","DOIUrl":"10.1186/s12935-024-03390-6","url":null,"abstract":"<p><p>Lung cancer (LC) ranks second most prevalent cancer in females after breast cancer and second in males after prostate cancer. Based on the GLOBOCAN 2020 report, India represented 5.9% of LC cases and 8.1% of deaths caused by the disease. Several clinical studies have shown that LC occurs because of biological and morphological abnormalities and the involvement of altered level of antioxidants, cytokines, and apoptotic markers. In the present study, we explored the antiproliferative activity of indeno[1,2-d]thiazolo[3,2-a]pyrimidine analogues against LC using in-vitro, in-silico, and in-vivo models. In-vitro screening against A549 cells revealed compounds 9B (8-methoxy-5-(3,4,5-trimethoxyphenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine) and 12B (5-(4-chlorophenyl)-5,6-dihydroindeno[1,2-d]thiazolo[3,2-a]pyrimidine) as potential pyrimidine analogues against LC. Compounds 9B and 12B were docked with different molecular targets IL-6, Cyt-C, Caspase9, and Caspase3 using AutoDock Vina 4.1 to evaluate the binding affinity. Subsequently, in-vivo studies were conducted in albino Wistar rats through ethyl-carbamate (EC)- induced LC. 9B and 12B imparted significant effects on physiological (weight variation), and biochemical (anti-oxidant [TBAR's, SOD, ProC, and GSH), lipid (TC, TG, LDL, VLDL, and HDL)], and cytokine (IL-2, IL-6, IL-10, and IL-1β) markers in EC-induced LC in albino Wistar rats. Morphological examination (SEM and H&E) and western blotting (IL-6, STAT3, Cyt-C, BAX, Bcl-2, Caspase3, and caspase9) showed that compounds 9B and 12B had antiproliferative effects. Accordingly, from the in-vitro, in-silico, and in-vivo experimental findings, we concluded that 9B and 12B have significant antiproliferative potential and are potential candidates for further evaluation to meet the requirements of investigation of new drug application.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11201866/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141455460","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Monitoring measurable residual disease in paediatric acute lymphoblastic leukaemia using immunoglobulin gene clonality based on next-generation sequencing. 利用基于新一代测序的免疫球蛋白基因克隆监测儿科急性淋巴细胞白血病的可测量残留疾病。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-25 DOI: 10.1186/s12935-024-03404-3
Won Kee Ahn, Kyunghee Yu, Hongkyung Kim, Seung-Tae Lee, Jong Rak Choi, Jung Woo Han, Chuhl Joo Lyu, Seungmin Hahn, Saeam Shin

Background: Assessment of measurable residual disease (MRD) is an essential prognostic tool for B-lymphoblastic leukaemia (B-ALL). In this study, we evaluated the utility of next-generation sequencing (NGS)-based MRD assessment in real-world clinical practice.

Method: The study included 93 paediatric patients with B-ALL treated at our institution between January 2017 and June 2022. Clonality for IGH or IGK rearrangements was identified in most bone marrow samples (91/93, 97.8%) obtained at diagnosis.

Results: In 421 monitoring samples, concordance was 74.8% between NGS and multiparameter flow cytometry and 70.7% between NGS and reverse transcription-PCR. Elevated quantities of clones of IGH alone (P < 0.001; hazard ratio [HR], 22.2; 95% confidence interval [CI], 7.1-69.1), IGK alone (P = 0.011; HR, 5.8; 95% CI, 1.5-22.5), and IGH or IGK (P < 0.001; HR, 7.2; 95% CI, 2.6-20.0) were associated with an increased risk of relapse. Detection of new clone(s) in NGS was also associated with inferior relapse-free survival (P < 0.001; HR, 18.1; 95% CI, 3.0-108.6). Multivariable analysis confirmed age at diagnosis, BCR::ABL1-like mutation, TCF3::PBX1 mutation, and increased quantity of IGH or IGK clones during monitoring as unfavourable factors.

Conclusion: In conclusion, this study highlights the usefulness of NGS-based MRD as a routine assessment tool for prognostication of paediatric patients with B-ALL.

背景:评估可测量残留疾病(MRD)是乙型淋巴细胞白血病(B-ALL)的重要预后工具。在这项研究中,我们评估了基于新一代测序(NGS)的 MRD 评估在实际临床实践中的实用性:研究纳入了 2017 年 1 月至 2022 年 6 月期间在我院接受治疗的 93 例 B-ALL 儿科患者。在诊断时获得的大多数骨髓样本(91/93,97.8%)中确定了 IGH 或 IGK 重排的克隆性:在 421 份监测样本中,NGS 与多参数流式细胞术的一致性为 74.8%,NGS 与反转录-PCR 的一致性为 70.7%。仅 IGH 克隆的数量升高(P 结论:NGS 和多参数流式细胞术的一致性为 74.8%,NGS 和反转录-PCR 的一致性为 70.7%:总之,本研究强调了基于 NGS 的 MRD 作为常规评估工具对 B-ALL 儿科患者预后的实用性。
{"title":"Monitoring measurable residual disease in paediatric acute lymphoblastic leukaemia using immunoglobulin gene clonality based on next-generation sequencing.","authors":"Won Kee Ahn, Kyunghee Yu, Hongkyung Kim, Seung-Tae Lee, Jong Rak Choi, Jung Woo Han, Chuhl Joo Lyu, Seungmin Hahn, Saeam Shin","doi":"10.1186/s12935-024-03404-3","DOIUrl":"10.1186/s12935-024-03404-3","url":null,"abstract":"<p><strong>Background: </strong>Assessment of measurable residual disease (MRD) is an essential prognostic tool for B-lymphoblastic leukaemia (B-ALL). In this study, we evaluated the utility of next-generation sequencing (NGS)-based MRD assessment in real-world clinical practice.</p><p><strong>Method: </strong>The study included 93 paediatric patients with B-ALL treated at our institution between January 2017 and June 2022. Clonality for IGH or IGK rearrangements was identified in most bone marrow samples (91/93, 97.8%) obtained at diagnosis.</p><p><strong>Results: </strong>In 421 monitoring samples, concordance was 74.8% between NGS and multiparameter flow cytometry and 70.7% between NGS and reverse transcription-PCR. Elevated quantities of clones of IGH alone (P < 0.001; hazard ratio [HR], 22.2; 95% confidence interval [CI], 7.1-69.1), IGK alone (P = 0.011; HR, 5.8; 95% CI, 1.5-22.5), and IGH or IGK (P < 0.001; HR, 7.2; 95% CI, 2.6-20.0) were associated with an increased risk of relapse. Detection of new clone(s) in NGS was also associated with inferior relapse-free survival (P < 0.001; HR, 18.1; 95% CI, 3.0-108.6). Multivariable analysis confirmed age at diagnosis, BCR::ABL1-like mutation, TCF3::PBX1 mutation, and increased quantity of IGH or IGK clones during monitoring as unfavourable factors.</p><p><strong>Conclusion: </strong>In conclusion, this study highlights the usefulness of NGS-based MRD as a routine assessment tool for prognostication of paediatric patients with B-ALL.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11201849/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141449806","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","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 sensitivity of acute myeloid leukemia cells to cytarabine is increased by suppressing the expression of Heme oxygenase-1 and hypoxia-inducible factor 1-alpha. 通过抑制血红素加氧酶-1 和缺氧诱导因子 1-α 的表达,可提高急性髓性白血病细胞对阿糖胞苷的敏感性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-06-25 DOI: 10.1186/s12935-024-03393-3
Mohammad Sadeghi, Asma Moslehi, Hadiseh Kheiry, Fariba Karoon Kiani, Asieh Zarei, Atefeh Khodakarami, Vahid Karpisheh, Ali Masjedi, Badrossadat Rahnama, Mohammad Hojjat-Farsangi, Mortaza Raeisi, Mehdi Yousefi, Ali Akbar Movasaghpour Akbari, Farhad Jadidi-Niaragh

Background: Acute myeloid leukemia (AML), a malignancy Often resistant to common chemotherapy regimens (Cytarabine (Ara-c) + Daunorubicin (DNR)), is accompanied by frequent relapses. Many factors are involved in causing chemoresistance. Heme Oxygenase-1 (HO-1) and Hypoxia-Inducible Factor 1-alpha (HIF-1α) are two of the most well-known genes, reported to be overexpressed in AML and promote resistance against chemotherapy according to several studies. The main chemotherapy agent used for AML treatment is Ara-c. We hypothesized that simultaneous targeting of HO-1 and HIF-1α could sensitize AML cells to Ara-c.

Method: In this study, we used our recently developed, Trans-Activator of Transcription (TAT) - Chitosan-Carboxymethyl Dextran (CCMD) - Poly Ethylene Glycol (PEG) - Nanoparticles (NPs), to deliver Ara-c along with siRNA molecules against the HO-1 and HIF-1α genes to AML primary cells (ex vivo) and cell lines including THP-1, KG-1, and HL-60 (in vitro). Subsequently, the effect of the single or combinational treatment on the growth, proliferation, apoptosis, and Reactive Oxygen Species (ROS) formation was evaluated.

Results: The designed NPs had a high potential in transfecting cells with siRNAs and drug. The results demonstrated that treatment of cells with Ara-c elevated the generation of ROS in the cells while decreasing the proliferation potential. Following the silencing of HO-1, the rate of apoptosis and ROS generation in response to Ara-c increased significantly. While proliferation and growth inhibition were considerably evident in HIF-1α-siRNA-transfected-AML cells compared to cells treated with free Ara-c. We found that the co-inhibition of genes could further sensitize AML cells to Ara-c treatment.

Conclusions: As far as we are aware, this study is the first to simultaneously inhibit the HO-1 and HIF-1α genes in AML using NPs. It can be concluded that HO-1 causes chemoresistance by protecting cells from ROS damage. Whereas, HIF-1α mostly exerts prolific and direct anti-apoptotic effects. These findings imply that simultaneous inhibition of HO-1 and HIF-1α can overcome Ara-c resistance and help improve the prognosis of AML patients.

背景:急性髓性白血病(AML急性髓性白血病(AML)是一种常对普通化疗方案(阿糖胞苷(Ara-c)+多柔比星(DNR))产生耐药性的恶性肿瘤,经常复发。导致化疗耐药性的因素有很多。血红素加氧酶-1(HO-1)和缺氧诱导因子 1-α(HIF-1α)是其中两个最著名的基因,多项研究表明,它们在急性髓细胞性白血病中过度表达,并促进化疗耐药性的产生。治疗急性髓细胞白血病的主要化疗药物是 Ara-c。我们假设,同时靶向 HO-1 和 HIF-1α 可使 AML 细胞对 Ara-c 敏感:在这项研究中,我们使用最近开发的转录激活剂(TAT)-壳聚糖-羧甲基葡聚糖(CCMD)-聚乙二醇(PEG)-纳米颗粒(NPs),将Ara-c与针对HO-1和HIF-1α基因的siRNA分子一起递送到AML原代细胞(体内外)和细胞系(体外),包括THP-1、KG-1和HL-60。随后,评估了单一或组合处理对细胞生长、增殖、凋亡和活性氧(ROS)形成的影响:结果:所设计的 NPs 在用 siRNAs 和药物转染细胞方面具有很高的潜力。结果表明,用 Ara-c 处理细胞会增加细胞中 ROS 的生成,同时降低细胞的增殖潜力。沉默HO-1后,细胞对Ara-c的凋亡率和ROS生成率显著增加。与用游离 Ara-c 处理的细胞相比,HIF-1α-siRNA 转染的AML 细胞的增殖和生长抑制更为明显。我们发现,基因的联合抑制可使 AML 细胞对 Ara-c 处理进一步敏感:据我们所知,这项研究是首次利用 NPs 同时抑制 AML 中的 HO-1 和 HIF-1α 基因。可以得出结论:HO-1通过保护细胞免受ROS损伤而导致化疗抵抗。而 HIF-1α 则主要发挥增殖和直接抗凋亡作用。这些发现意味着,同时抑制HO-1和HIF-1α可以克服Ara-c耐药性,有助于改善急性髓细胞白血病患者的预后。
{"title":"The sensitivity of acute myeloid leukemia cells to cytarabine is increased by suppressing the expression of Heme oxygenase-1 and hypoxia-inducible factor 1-alpha.","authors":"Mohammad Sadeghi, Asma Moslehi, Hadiseh Kheiry, Fariba Karoon Kiani, Asieh Zarei, Atefeh Khodakarami, Vahid Karpisheh, Ali Masjedi, Badrossadat Rahnama, Mohammad Hojjat-Farsangi, Mortaza Raeisi, Mehdi Yousefi, Ali Akbar Movasaghpour Akbari, Farhad Jadidi-Niaragh","doi":"10.1186/s12935-024-03393-3","DOIUrl":"10.1186/s12935-024-03393-3","url":null,"abstract":"<p><strong>Background: </strong>Acute myeloid leukemia (AML), a malignancy Often resistant to common chemotherapy regimens (Cytarabine (Ara-c) + Daunorubicin (DNR)), is accompanied by frequent relapses. Many factors are involved in causing chemoresistance. Heme Oxygenase-1 (HO-1) and Hypoxia-Inducible Factor 1-alpha (HIF-1α) are two of the most well-known genes, reported to be overexpressed in AML and promote resistance against chemotherapy according to several studies. The main chemotherapy agent used for AML treatment is Ara-c. We hypothesized that simultaneous targeting of HO-1 and HIF-1α could sensitize AML cells to Ara-c.</p><p><strong>Method: </strong>In this study, we used our recently developed, Trans-Activator of Transcription (TAT) - Chitosan-Carboxymethyl Dextran (CCMD) - Poly Ethylene Glycol (PEG) - Nanoparticles (NPs), to deliver Ara-c along with siRNA molecules against the HO-1 and HIF-1α genes to AML primary cells (ex vivo) and cell lines including THP-1, KG-1, and HL-60 (in vitro). Subsequently, the effect of the single or combinational treatment on the growth, proliferation, apoptosis, and Reactive Oxygen Species (ROS) formation was evaluated.</p><p><strong>Results: </strong>The designed NPs had a high potential in transfecting cells with siRNAs and drug. The results demonstrated that treatment of cells with Ara-c elevated the generation of ROS in the cells while decreasing the proliferation potential. Following the silencing of HO-1, the rate of apoptosis and ROS generation in response to Ara-c increased significantly. While proliferation and growth inhibition were considerably evident in HIF-1α-siRNA-transfected-AML cells compared to cells treated with free Ara-c. We found that the co-inhibition of genes could further sensitize AML cells to Ara-c treatment.</p><p><strong>Conclusions: </strong>As far as we are aware, this study is the first to simultaneously inhibit the HO-1 and HIF-1α genes in AML using NPs. It can be concluded that HO-1 causes chemoresistance by protecting cells from ROS damage. Whereas, HIF-1α mostly exerts prolific and direct anti-apoptotic effects. These findings imply that simultaneous inhibition of HO-1 and HIF-1α can overcome Ara-c resistance and help improve the prognosis of AML patients.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11197338/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141449807","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","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
Cancer Cell International
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