首页 > 最新文献

Cancer Cell International最新文献

英文 中文
PRRX1-OLR1 axis supports CAFs-mediated lung cancer progression and immune suppression. PRRX1-OLR1 轴支持 CAFs 介导的肺癌进展和免疫抑制。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-15 DOI: 10.1186/s12935-024-03436-9
Yunhao Sun, Kaijun Ying, Jian Sun, Yao Wang, Limin Qiu, Mingming Ji, Lin Sun, Jinjin Chen

Objective: To investigate the mechanism by which cancer-associated fibroblasts (CAFs) affect the growth and immune evasion of lung cancer cells.

Methods: Initially, datasets comparing CAFs with normal fibroblasts were downloaded from the GEO dataset GSE48397. Genes with the most significant differential expression were selected and validated using clinical data. Subsequently, CAFs were isolated, and the selected genes were knocked down in CAFs. Co-culture experiments were conducted with H1299 or A549 cells to analyze changes in lung cancer cell growth, migration, and immune evasion in vitro and in vivo. To further elucidate the upstream regulatory mechanism, relevant ChIP-seq data were downloaded from the GEO database, and the regulatory relationships were validated through ChIP-qPCR and luciferase reporter assays.

Results: OLR1 was significantly overexpressed in CAFs and strongly correlated with adverse prognosis in lung cancer patients. Knockdown of OLR1 markedly inhibited CAFs' support for the growth and immune evasion of lung cancer cells in vitro and in vivo. ChIP-seq results demonstrated that PRRX1 can promote OLR1 expression by recruiting H3K27ac and H3K4me3, thereby activating CAFs. Knockdown of PRRX1 significantly inhibited CAFs' function, while further overexpression of OLR1 restored CAFs' support for lung cancer cell growth, migration, and immune evasion.

Conclusion: PRRX1 promotes OLR1 expression by recruiting H3K27ac and H3K4me3, activating CAFs, and thereby promoting the growth, migration, and immune evasion of lung cancer cells.

目的:研究癌症相关成纤维细胞(CAFs)影响肺癌细胞生长和免疫逃避的机制:研究癌症相关成纤维细胞(CAFs)影响肺癌细胞生长和免疫逃避的机制:首先,从 GEO 数据集 GSE48397 中下载比较 CAF 与正常成纤维细胞的数据集。筛选出差异表达最明显的基因,并通过临床数据进行验证。随后,分离出 CAFs,并在 CAFs 中敲除所选基因。与 H1299 或 A549 细胞进行共培养实验,分析肺癌细胞在体外和体内的生长、迁移和免疫逃避变化。为了进一步阐明上游调控机制,研究人员从 GEO 数据库中下载了相关的 ChIP-seq 数据,并通过 ChIP-qPCR 和荧光素酶报告实验验证了调控关系:结果:OLR1在CAFs中明显过表达,并与肺癌患者的不良预后密切相关。敲除 OLR1 能明显抑制 CAFs 在体外和体内对肺癌细胞生长和免疫逃避的支持。ChIP-seq结果表明,PRRX1可通过招募H3K27ac和H3K4me3促进OLR1的表达,从而激活CAFs。敲除PRRX1可显著抑制CAFs的功能,而进一步过表达OLR1可恢复CAFs对肺癌细胞生长、迁移和免疫逃避的支持:结论:PRRX1通过招募H3K27ac和H3K4me3促进OLR1的表达,激活CAFs,从而促进肺癌细胞的生长、迁移和免疫逃避。
{"title":"PRRX1-OLR1 axis supports CAFs-mediated lung cancer progression and immune suppression.","authors":"Yunhao Sun, Kaijun Ying, Jian Sun, Yao Wang, Limin Qiu, Mingming Ji, Lin Sun, Jinjin Chen","doi":"10.1186/s12935-024-03436-9","DOIUrl":"10.1186/s12935-024-03436-9","url":null,"abstract":"<p><strong>Objective: </strong>To investigate the mechanism by which cancer-associated fibroblasts (CAFs) affect the growth and immune evasion of lung cancer cells.</p><p><strong>Methods: </strong>Initially, datasets comparing CAFs with normal fibroblasts were downloaded from the GEO dataset GSE48397. Genes with the most significant differential expression were selected and validated using clinical data. Subsequently, CAFs were isolated, and the selected genes were knocked down in CAFs. Co-culture experiments were conducted with H1299 or A549 cells to analyze changes in lung cancer cell growth, migration, and immune evasion in vitro and in vivo. To further elucidate the upstream regulatory mechanism, relevant ChIP-seq data were downloaded from the GEO database, and the regulatory relationships were validated through ChIP-qPCR and luciferase reporter assays.</p><p><strong>Results: </strong>OLR1 was significantly overexpressed in CAFs and strongly correlated with adverse prognosis in lung cancer patients. Knockdown of OLR1 markedly inhibited CAFs' support for the growth and immune evasion of lung cancer cells in vitro and in vivo. ChIP-seq results demonstrated that PRRX1 can promote OLR1 expression by recruiting H3K27ac and H3K4me3, thereby activating CAFs. Knockdown of PRRX1 significantly inhibited CAFs' function, while further overexpression of OLR1 restored CAFs' support for lung cancer cell growth, migration, and immune evasion.</p><p><strong>Conclusion: </strong>PRRX1 promotes OLR1 expression by recruiting H3K27ac and H3K4me3, activating CAFs, and thereby promoting the growth, migration, and immune evasion of lung cancer cells.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11251326/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619402","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
Retraction Note: LncRNA LINC00337 sponges mir-1285-3p to promote proliferation and metastasis of lung adenocarcinoma cells by upregulating YTHDF1. 撤稿说明:LncRNA LINC00337通过上调YTHDF1海绵化mir-1285-3p,促进肺腺癌细胞的增殖和转移。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-14 DOI: 10.1186/s12935-024-03431-0
Ru-Nan Zhang, Dong-Mei Wu, Li-Ping Wu, Guo-Wei Gao
{"title":"Retraction Note: LncRNA LINC00337 sponges mir-1285-3p to promote proliferation and metastasis of lung adenocarcinoma cells by upregulating YTHDF1.","authors":"Ru-Nan Zhang, Dong-Mei Wu, Li-Ping Wu, Guo-Wei Gao","doi":"10.1186/s12935-024-03431-0","DOIUrl":"10.1186/s12935-024-03431-0","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247831/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141615953","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
Key genes and molecular mechanisms related to Paclitaxel Resistance. 与紫杉醇耐药性相关的关键基因和分子机制。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-13 DOI: 10.1186/s12935-024-03415-0
Adel I Alalawy

Paclitaxel is commonly used to treat breast, ovarian, lung, esophageal, gastric, pancreatic cancer, and neck cancer cells. Cancer recurrence is observed in patients treated with paclitaxel due to paclitaxel resistance emergence. Resistant mechanisms are observed in cancer cells treated with paclitaxel, docetaxel, and cabazitaxel including changes in the target molecule β-tubulin of mitosis, molecular mechanisms that activate efflux drug out of the cells, and alterations in regulatory proteins of apoptosis. This review discusses new molecular mechanisms of taxane resistance, such as overexpression of genes like the multidrug resistance genes and EDIL3, ABCB1, MRP1, and TRAG-3/CSAG2 genes. Moreover, significant lncRNAs are detected in paclitaxel resistance, such as lncRNA H19 and cross-resistance between taxanes. This review contributed to discovering new treatment strategies for taxane resistance and increasing the responsiveness of cancer cells toward chemotherapeutic drugs.

紫杉醇常用于治疗乳腺癌、卵巢癌、肺癌、食道癌、胃癌、胰腺癌和颈部癌细胞。接受紫杉醇治疗的患者由于出现紫杉醇抗药性而导致癌症复发。在接受紫杉醇、多西他赛和卡巴齐他赛治疗的癌细胞中观察到了抗药性机制,包括有丝分裂靶分子β-tubulin的变化、激活药物流出细胞的分子机制以及细胞凋亡调节蛋白的变化。本综述讨论了紫杉类药物耐药性的新分子机制,如多药耐药基因、EDIL3、ABCB1、MRP1 和 TRAG-3/CSAG2 等基因的过度表达。此外,在紫杉醇耐药性中发现了重要的lncRNA,如lncRNA H19和紫杉醇类药物之间的交叉耐药性。这篇综述有助于发现新的紫杉类药物耐药性治疗策略,提高癌细胞对化疗药物的反应性。
{"title":"Key genes and molecular mechanisms related to Paclitaxel Resistance.","authors":"Adel I Alalawy","doi":"10.1186/s12935-024-03415-0","DOIUrl":"10.1186/s12935-024-03415-0","url":null,"abstract":"<p><p>Paclitaxel is commonly used to treat breast, ovarian, lung, esophageal, gastric, pancreatic cancer, and neck cancer cells. Cancer recurrence is observed in patients treated with paclitaxel due to paclitaxel resistance emergence. Resistant mechanisms are observed in cancer cells treated with paclitaxel, docetaxel, and cabazitaxel including changes in the target molecule β-tubulin of mitosis, molecular mechanisms that activate efflux drug out of the cells, and alterations in regulatory proteins of apoptosis. This review discusses new molecular mechanisms of taxane resistance, such as overexpression of genes like the multidrug resistance genes and EDIL3, ABCB1, MRP1, and TRAG-3/CSAG2 genes. Moreover, significant lncRNAs are detected in paclitaxel resistance, such as lncRNA H19 and cross-resistance between taxanes. This review contributed to discovering new treatment strategies for taxane resistance and increasing the responsiveness of cancer cells toward chemotherapeutic drugs.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11245874/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141603308","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
GLP and G9a histone methyltransferases as potential therapeutic targets for lymphoid neoplasms. 作为淋巴肿瘤潜在治疗靶点的 GLP 和 G9a 组蛋白甲基转移酶。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-12 DOI: 10.1186/s12935-024-03441-y
Amandda Évelin Silva-Carvalho, Luma Dayane Carvalho Filiú-Braga, Gabriela Muller Reche Bogéa, Alan Jhones Barbosa de Assis, Fábio Pittella-Silva, Felipe Saldanha-Araujo

Histone methyltransferases (HMTs) are enzymes that regulate histone methylation and play an important role in controlling transcription by altering the chromatin structure. Aberrant activation of HMTs has been widely reported in certain types of neoplastic cells. Among them, G9a/EHMT2 and GLP/EHMT1 are crucial for H3K9 methylation, and their dysregulation has been associated with tumor initiation and progression in different types of cancer. More recently, it has been shown that G9a and GLP appear to play a critical role in several lymphoid hematologic malignancies. Importantly, the key roles played by both enzymes in various diseases made them attractive targets for drug development. In fact, in recent years, several groups have tried to develop small molecule inhibitors targeting their epigenetic activities as potential anticancer therapeutic tools. In this review, we discuss the physiological role of GLP and G9a, their oncogenic functions in hematologic malignancies of the lymphoid lineage, and the therapeutic potential of epigenetic drugs targeting G9a/GLP for cancer treatment.

组蛋白甲基转移酶(HMTs)是一种调节组蛋白甲基化的酶,通过改变染色质结构在控制转录方面发挥着重要作用。在某些类型的肿瘤细胞中,HMTs 的异常激活已被广泛报道。其中,G9a/EHMT2 和 GLP/EHMT1 对 H3K9 甲基化至关重要,它们的失调与不同类型癌症的肿瘤发生和发展有关。最近的研究表明,G9a 和 GLP 似乎在几种淋巴血液恶性肿瘤中发挥着关键作用。重要的是,这两种酶在各种疾病中发挥的关键作用使它们成为具有吸引力的药物开发目标。事实上,近年来已有多个研究小组尝试开发针对其表观遗传活性的小分子抑制剂,作为潜在的抗癌治疗工具。在这篇综述中,我们将讨论 GLP 和 G9a 的生理作用、它们在淋巴系血液恶性肿瘤中的致癌功能,以及针对 G9a/GLP 的表观遗传药物在癌症治疗中的潜力。
{"title":"GLP and G9a histone methyltransferases as potential therapeutic targets for lymphoid neoplasms.","authors":"Amandda Évelin Silva-Carvalho, Luma Dayane Carvalho Filiú-Braga, Gabriela Muller Reche Bogéa, Alan Jhones Barbosa de Assis, Fábio Pittella-Silva, Felipe Saldanha-Araujo","doi":"10.1186/s12935-024-03441-y","DOIUrl":"10.1186/s12935-024-03441-y","url":null,"abstract":"<p><p>Histone methyltransferases (HMTs) are enzymes that regulate histone methylation and play an important role in controlling transcription by altering the chromatin structure. Aberrant activation of HMTs has been widely reported in certain types of neoplastic cells. Among them, G9a/EHMT2 and GLP/EHMT1 are crucial for H3K9 methylation, and their dysregulation has been associated with tumor initiation and progression in different types of cancer. More recently, it has been shown that G9a and GLP appear to play a critical role in several lymphoid hematologic malignancies. Importantly, the key roles played by both enzymes in various diseases made them attractive targets for drug development. In fact, in recent years, several groups have tried to develop small molecule inhibitors targeting their epigenetic activities as potential anticancer therapeutic tools. In this review, we discuss the physiological role of GLP and G9a, their oncogenic functions in hematologic malignancies of the lymphoid lineage, and the therapeutic potential of epigenetic drugs targeting G9a/GLP for cancer treatment.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11249034/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141598533","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
Enhancing cancer therapy: the integration of oncolytic virus therapy with diverse treatments 加强癌症治疗:溶瘤病毒疗法与多种治疗方法的结合
IF 5.8 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-11 DOI: 10.1186/s12935-024-03424-z
Zhuo Yan, Zhengbo Zhang, Yanan Chen, Jianghua Xu, Jilong Wang, Zhangquan Wang
As one of the significant challenges to human health, cancer has long been a focal point in medical treatment. With ongoing advancements in the field of medicine, numerous methodologies for cancer therapy have emerged, among which oncolytic virus therapy has gained considerable attention. However, oncolytic viruses still exhibit limitations. Combining them with various therapies can further enhance the efficacy of cancer treatment, offering renewed hope for patients. In recent research, scientists have recognized the promising prospect of amalgamating oncolytic virus therapy with diverse treatments, potentially surmounting the restrictions of singular approaches. The central concept of this combined therapy revolves around leveraging oncolytic virus to incite localized tumor inflammation, augmenting the immune response for immunotherapeutic efficacy. Through this approach, the patient's immune system can better recognize and eliminate cancer cells, simultaneously reducing tumor evasion mechanisms against the immune system. This review delves deeply into the latest research progress concerning the integration of oncolytic virus with diverse treatments and its role in various types of cancer therapy. We aim to analyze the mechanisms, advantages, potential challenges, and future research directions of this combination therapy. By extensively exploring this field, we aim to instill renewed hope in the fight against cancer.
癌症是人类健康面临的重大挑战之一,长期以来一直是医学治疗的焦点。随着医学领域的不断进步,出现了许多治疗癌症的方法,其中溶瘤病毒疗法备受关注。然而,溶瘤病毒仍有其局限性。将它们与各种疗法相结合,可以进一步提高癌症治疗的疗效,为患者带来新的希望。在最近的研究中,科学家们认识到,将溶瘤病毒疗法与多种疗法相结合,有可能突破单一疗法的限制,前景十分广阔。这种联合疗法的核心理念是利用溶瘤病毒激发局部肿瘤炎症,增强免疫反应以达到免疫治疗效果。通过这种方法,患者的免疫系统可以更好地识别和消灭癌细胞,同时减少肿瘤逃避免疫系统的机制。本综述深入探讨了溶瘤病毒与多种治疗方法相结合的最新研究进展及其在各类癌症治疗中的作用。我们旨在分析这种联合疗法的机制、优势、潜在挑战和未来研究方向。通过对这一领域的广泛探索,我们希望为抗击癌症带来新的希望。
{"title":"Enhancing cancer therapy: the integration of oncolytic virus therapy with diverse treatments","authors":"Zhuo Yan, Zhengbo Zhang, Yanan Chen, Jianghua Xu, Jilong Wang, Zhangquan Wang","doi":"10.1186/s12935-024-03424-z","DOIUrl":"https://doi.org/10.1186/s12935-024-03424-z","url":null,"abstract":"As one of the significant challenges to human health, cancer has long been a focal point in medical treatment. With ongoing advancements in the field of medicine, numerous methodologies for cancer therapy have emerged, among which oncolytic virus therapy has gained considerable attention. However, oncolytic viruses still exhibit limitations. Combining them with various therapies can further enhance the efficacy of cancer treatment, offering renewed hope for patients. In recent research, scientists have recognized the promising prospect of amalgamating oncolytic virus therapy with diverse treatments, potentially surmounting the restrictions of singular approaches. The central concept of this combined therapy revolves around leveraging oncolytic virus to incite localized tumor inflammation, augmenting the immune response for immunotherapeutic efficacy. Through this approach, the patient's immune system can better recognize and eliminate cancer cells, simultaneously reducing tumor evasion mechanisms against the immune system. This review delves deeply into the latest research progress concerning the integration of oncolytic virus with diverse treatments and its role in various types of cancer therapy. We aim to analyze the mechanisms, advantages, potential challenges, and future research directions of this combination therapy. By extensively exploring this field, we aim to instill renewed hope in the fight against cancer.","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.8,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141585226","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inhibition of autophagy induced by tetrandrine promotes the accumulation of reactive oxygen species and sensitizes efficacy of tetrandrine in pancreatic cancer. 抑制四氢嘌呤诱导的自噬会促进活性氧的积累,并削弱四氢嘌呤对胰腺癌的疗效。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-10 DOI: 10.1186/s12935-024-03410-5
Yiwei Wang, Ting Xu, Hongcheng Wang, Guanggai Xia, Xinyu Huang

Pancreatic cancer, characterized by its poor prognosis, exhibits a marked resistance to conventional chemotherapy and immunotherapy, underscoring the urgent need for more effective treatment modalities. In light of this, the present study is designed to assess the potential antineoplastic efficacy of a combined regimen involving tetrandrine, a plant-derived alkaloid, and autophagy inhibitors in the context of pancreatic cancer. Electron microscopy and immunoblots showed that tetrandrine promoted the formation of autophagosomes and the upregulation of LC3II and the downregulation of p62 expression, indicating that tetrandrine induced autophagy in pancreatic cancer cells. Western blot revealed that tetrandrine inhibited the phosphorylation of AKT and mTOR, as well as the expression of Bcl-2, while upregulating Beclin-1 expression. Moreover, tetrandrine promoted the transcription and protein expression of ATG7. Following the combination of autophagy inhibitors and tetrandrine, the apoptotic rate and cell death significantly increased in pancreatic cancer cells. Consistent results were obtained when ATG7 was silenced. Additionally, tetrandrine induced the generation of ROS, which was involved in the activation of autophagy and apoptosis. Further investigation revealed that upon autophagy inhibition, ROS accumulated in pancreatic cancer cells, resulting in decreased mitochondrial membrane potential and further induction of apoptosis. The results of treating subcutaneous xenograft tumors with a combination of tetrandrine and chloroquine validated that autophagy inhibition enhances the toxicity of tetrandrine against pancreatic cancer in vivo. Altogether, our study demonstrates that tetrandrine induces cytoprotective autophagy in pancreatic cancer cells. Inhibiting tetrandrine-induced autophagy promotes the accumulation of ROS and enhances its toxicity against pancreatic cancer.

胰腺癌的特点是预后不良,对传统化疗和免疫疗法表现出明显的抗药性,因此迫切需要更有效的治疗方法。有鉴于此,本研究旨在评估在胰腺癌的治疗中,四氢化可的松(一种植物提取的生物碱)和自噬抑制剂联合疗法的潜在抗肿瘤疗效。电子显微镜和免疫印迹显示,四氢化可促进自噬体的形成、LC3II的上调和p62表达的下调,表明四氢化可诱导胰腺癌细胞的自噬。Western 印迹显示,四氢化苦参碱抑制了 AKT 和 mTOR 的磷酸化以及 Bcl-2 的表达,同时上调了 Beclin-1 的表达。此外,四氢嘌呤还能促进 ATG7 的转录和蛋白表达。自噬抑制剂和四氢化甘油联合使用后,胰腺癌细胞的凋亡率和细胞死亡率显著增加。在沉默 ATG7 的情况下也得到了一致的结果。此外,四氢化可诱导产生 ROS,而 ROS 参与了自噬和细胞凋亡的激活。进一步研究发现,抑制自噬后,胰腺癌细胞中的 ROS 会累积,导致线粒体膜电位降低,进一步诱导细胞凋亡。用四氢蒽醌和氯喹联合治疗皮下异种移植瘤的结果验证了自噬抑制可增强四氢蒽醌对胰腺癌的体内毒性。总之,我们的研究表明,四氢化可诱导胰腺癌细胞中具有细胞保护作用的自噬。抑制四肾上腺素诱导的自噬作用会促进 ROS 的积累,并增强其对胰腺癌的毒性。
{"title":"Inhibition of autophagy induced by tetrandrine promotes the accumulation of reactive oxygen species and sensitizes efficacy of tetrandrine in pancreatic cancer.","authors":"Yiwei Wang, Ting Xu, Hongcheng Wang, Guanggai Xia, Xinyu Huang","doi":"10.1186/s12935-024-03410-5","DOIUrl":"10.1186/s12935-024-03410-5","url":null,"abstract":"<p><p>Pancreatic cancer, characterized by its poor prognosis, exhibits a marked resistance to conventional chemotherapy and immunotherapy, underscoring the urgent need for more effective treatment modalities. In light of this, the present study is designed to assess the potential antineoplastic efficacy of a combined regimen involving tetrandrine, a plant-derived alkaloid, and autophagy inhibitors in the context of pancreatic cancer. Electron microscopy and immunoblots showed that tetrandrine promoted the formation of autophagosomes and the upregulation of LC3II and the downregulation of p62 expression, indicating that tetrandrine induced autophagy in pancreatic cancer cells. Western blot revealed that tetrandrine inhibited the phosphorylation of AKT and mTOR, as well as the expression of Bcl-2, while upregulating Beclin-1 expression. Moreover, tetrandrine promoted the transcription and protein expression of ATG7. Following the combination of autophagy inhibitors and tetrandrine, the apoptotic rate and cell death significantly increased in pancreatic cancer cells. Consistent results were obtained when ATG7 was silenced. Additionally, tetrandrine induced the generation of ROS, which was involved in the activation of autophagy and apoptosis. Further investigation revealed that upon autophagy inhibition, ROS accumulated in pancreatic cancer cells, resulting in decreased mitochondrial membrane potential and further induction of apoptosis. The results of treating subcutaneous xenograft tumors with a combination of tetrandrine and chloroquine validated that autophagy inhibition enhances the toxicity of tetrandrine against pancreatic cancer in vivo. Altogether, our study demonstrates that tetrandrine induces cytoprotective autophagy in pancreatic cancer cells. Inhibiting tetrandrine-induced autophagy promotes the accumulation of ROS and enhances its toxicity against pancreatic cancer.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11238362/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141579040","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 common bisulfite-conversion-based techniques to analyze DNA methylation in human cancers. 基于亚硫酸氢盐转换的常用技术,用于分析人类癌症中的 DNA 甲基化。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-09 DOI: 10.1186/s12935-024-03405-2
Farhad Jeddi, Elnaz Faghfuri, Sahar Mehranfar, Narges Soozangar

DNA methylation is an important molecular modification that plays a key role in the expression of cancer genes. Evaluation of epigenetic changes, hypomethylation and hypermethylation, in specific genes are applied for cancer diagnosis. Numerous studies have concentrated on describing DNA methylation patterns as biomarkers for cancer diagnosis monitoring and predicting response to cancer therapy. Various techniques for detecting DNA methylation status in cancers are based on sodium bisulfite treatment. According to the application of these methods in research and clinical studies, they have a number of advantages and disadvantages. The current review highlights sodium bisulfite treatment-based techniques, as well as, the advantages, drawbacks, and applications of these methods in the evaluation of human cancers.

DNA 甲基化是一种重要的分子修饰,在癌症基因的表达中起着关键作用。评估特定基因的表观遗传变化(低甲基化和高甲基化)可用于癌症诊断。大量研究集中于描述 DNA 甲基化模式,将其作为癌症诊断监测和预测癌症治疗反应的生物标志物。各种检测癌症 DNA 甲基化状态的技术都是基于亚硫酸氢钠处理法。根据这些方法在研究和临床研究中的应用情况,它们各有利弊。本综述重点介绍了基于亚硫酸氢钠处理的技术,以及这些方法的优点、缺点和在人类癌症评估中的应用。
{"title":"The common bisulfite-conversion-based techniques to analyze DNA methylation in human cancers.","authors":"Farhad Jeddi, Elnaz Faghfuri, Sahar Mehranfar, Narges Soozangar","doi":"10.1186/s12935-024-03405-2","DOIUrl":"10.1186/s12935-024-03405-2","url":null,"abstract":"<p><p>DNA methylation is an important molecular modification that plays a key role in the expression of cancer genes. Evaluation of epigenetic changes, hypomethylation and hypermethylation, in specific genes are applied for cancer diagnosis. Numerous studies have concentrated on describing DNA methylation patterns as biomarkers for cancer diagnosis monitoring and predicting response to cancer therapy. Various techniques for detecting DNA methylation status in cancers are based on sodium bisulfite treatment. According to the application of these methods in research and clinical studies, they have a number of advantages and disadvantages. The current review highlights sodium bisulfite treatment-based techniques, as well as, the advantages, drawbacks, and applications of these methods in the evaluation of human cancers.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11234524/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141562638","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
Promoting reactive oxygen species accumulation to overcome tyrosine kinase inhibitor resistance in cancer. 促进活性氧积累,克服癌症中酪氨酸激酶抑制剂的耐药性。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-09 DOI: 10.1186/s12935-024-03418-x
Wei Lin, Xiaojun Wang, Mingxin Diao, Yangwei Wang, Rong Zhao, Jiaping Chen, Yongde Liao, Qinghong Long, Yunchong Meng

Background: In tumor treatment, protein tyrosine kinase inhibitors (TKIs) have been extensively utilized. However, the efficacy of TKI is significantly compromised by drug resistance. Consequently, finding an effective solution to overcome TKI resistance becomes crucial. Reactive oxygen species (ROS) are a group of highly active molecules that play important roles in targeted cancer therapy including TKI targeted therapy. In this review, we concentrate on the ROS-associated mechanisms of TKI lethality in tumors and strategies for regulating ROS to reverse TKI resistance in cancer.

Main body: Elevated ROS levels often manifest during TKI therapy in cancers, potentially causing organelle damage and cell death, which are critical to the success of TKIs in eradicating cancer cells. However, it is noteworthy that cancer cells might initiate resistance pathways to shield themselves from ROS-induced damage, leading to TKI resistance. Addressing this challenge involves blocking these resistance pathways, for instance, the NRF2-KEAP1 axis and protective autophagy, to promote ROS accumulation in cells, thereby resensitizing drug-resistant cancer cells to TKIs. Additional effective approaches inducing ROS generation within drug-resistant cells and providing exogenous ROS stimulation.

Conclusion: ROS play pivotal roles in the eradication of tumor cells by TKI. Harnessing the accumulation of ROS to overcome TKI resistance is an effective and widely applicable approach.

背景:在肿瘤治疗中,蛋白酪氨酸激酶抑制剂(TKIs)得到了广泛应用。然而,TKI 的疗效因耐药性而大打折扣。因此,找到克服 TKI 耐药性的有效解决方案变得至关重要。活性氧(ROS)是一类高活性分子,在包括 TKI 靶向治疗在内的癌症靶向治疗中发挥着重要作用。在这篇综述中,我们将集中讨论肿瘤中 TKI 致死的 ROS 相关机制,以及调节 ROS 以逆转癌症中 TKI 耐药性的策略:癌症TKI治疗过程中经常会出现ROS水平升高,有可能导致细胞器损伤和细胞死亡,这对TKIs成功根除癌细胞至关重要。然而,值得注意的是,癌细胞可能会启动抗性途径来抵御 ROS 诱导的损伤,从而导致 TKI 抗性。要应对这一挑战,就必须阻断这些抵抗途径,例如 NRF2-KEAP1 轴和保护性自噬,以促进细胞中 ROS 的积累,从而使耐药癌细胞对 TKIs 再敏感。在耐药细胞内诱导 ROS 生成和提供外源性 ROS 刺激的其他有效方法:结论:ROS 在 TKIs 根除肿瘤细胞的过程中发挥着关键作用。利用 ROS 的积累来克服 TKI 耐药性是一种有效且广泛适用的方法。
{"title":"Promoting reactive oxygen species accumulation to overcome tyrosine kinase inhibitor resistance in cancer.","authors":"Wei Lin, Xiaojun Wang, Mingxin Diao, Yangwei Wang, Rong Zhao, Jiaping Chen, Yongde Liao, Qinghong Long, Yunchong Meng","doi":"10.1186/s12935-024-03418-x","DOIUrl":"10.1186/s12935-024-03418-x","url":null,"abstract":"<p><strong>Background: </strong>In tumor treatment, protein tyrosine kinase inhibitors (TKIs) have been extensively utilized. However, the efficacy of TKI is significantly compromised by drug resistance. Consequently, finding an effective solution to overcome TKI resistance becomes crucial. Reactive oxygen species (ROS) are a group of highly active molecules that play important roles in targeted cancer therapy including TKI targeted therapy. In this review, we concentrate on the ROS-associated mechanisms of TKI lethality in tumors and strategies for regulating ROS to reverse TKI resistance in cancer.</p><p><strong>Main body: </strong>Elevated ROS levels often manifest during TKI therapy in cancers, potentially causing organelle damage and cell death, which are critical to the success of TKIs in eradicating cancer cells. However, it is noteworthy that cancer cells might initiate resistance pathways to shield themselves from ROS-induced damage, leading to TKI resistance. Addressing this challenge involves blocking these resistance pathways, for instance, the NRF2-KEAP1 axis and protective autophagy, to promote ROS accumulation in cells, thereby resensitizing drug-resistant cancer cells to TKIs. Additional effective approaches inducing ROS generation within drug-resistant cells and providing exogenous ROS stimulation.</p><p><strong>Conclusion: </strong>ROS play pivotal roles in the eradication of tumor cells by TKI. Harnessing the accumulation of ROS to overcome TKI resistance is an effective and widely applicable approach.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11234736/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141562637","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
USP8 promotes the tumorigenesis of intrahepatic cholangiocarcinoma via stabilizing OGT. USP8 通过稳定 OGT 促进肝内胆管癌的肿瘤发生。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-07 DOI: 10.1186/s12935-024-03370-w
Guo Long, Dong Wang, Jianing Tang, Kuan Hu, Ledu Zhou

Ubiquitination was considered to be a crucial factor in intrahepatic cholangiocarcinoma (iCCA) development. Herein, we identified Ubiquitin-specific peptidase 8 (USP8) as a key regulator for promoting the tumorigenesis of iCCA cell via stabilizing OGT. USP8 was overexpressed in human tumor tissues and correlated with worse survival. Moreover, the mass spectrometry and co-immunoprecipitation analysis indicated that USP8 interacted with OGT. USP8 worked as a bona fide deubiquitylase of OGT. It stabilized OGT in a deubiquitylation activity-dependent manner. Meanwhile, DUB-IN3, the USP8 inhibitor, could also restrain the malignancy of intrahepatic cholangiocarcinoma. In addition, USP8 depletion promoted the response of iCCA to pemigatinib. In conclusion, our findings pointed to a previously undocumented catalytic role for USP8 as a deubiquitinating enzyme of OGT. The USP8-OGT axis could be a potential target for iCCA therapy.

泛素化被认为是肝内胆管癌(iCCA)发生的关键因素。在此,我们发现泛素特异性肽酶 8(USP8)是通过稳定 OGT 促进 iCCA 细胞肿瘤发生的关键调控因子。USP8 在人类肿瘤组织中过表达,并与生存率下降相关。此外,质谱分析和共沉淀分析表明,USP8 与 OGT 相互作用。USP8 是 OGT 的真正去泛素化酶。它以依赖于去泛素化活性的方式稳定 OGT。同时,USP8抑制剂DUB-IN3也能抑制肝内胆管癌的恶性发展。此外,USP8耗竭可促进iCCA对pemigatinib的反应。总之,我们的研究结果表明,USP8作为OGT的去泛素化酶,具有以前未曾记载的催化作用。USP8-OGT 轴可能是治疗 iCCA 的潜在靶点。
{"title":"USP8 promotes the tumorigenesis of intrahepatic cholangiocarcinoma via stabilizing OGT.","authors":"Guo Long, Dong Wang, Jianing Tang, Kuan Hu, Ledu Zhou","doi":"10.1186/s12935-024-03370-w","DOIUrl":"10.1186/s12935-024-03370-w","url":null,"abstract":"<p><p>Ubiquitination was considered to be a crucial factor in intrahepatic cholangiocarcinoma (iCCA) development. Herein, we identified Ubiquitin-specific peptidase 8 (USP8) as a key regulator for promoting the tumorigenesis of iCCA cell via stabilizing OGT. USP8 was overexpressed in human tumor tissues and correlated with worse survival. Moreover, the mass spectrometry and co-immunoprecipitation analysis indicated that USP8 interacted with OGT. USP8 worked as a bona fide deubiquitylase of OGT. It stabilized OGT in a deubiquitylation activity-dependent manner. Meanwhile, DUB-IN3, the USP8 inhibitor, could also restrain the malignancy of intrahepatic cholangiocarcinoma. In addition, USP8 depletion promoted the response of iCCA to pemigatinib. In conclusion, our findings pointed to a previously undocumented catalytic role for USP8 as a deubiquitinating enzyme of OGT. The USP8-OGT axis could be a potential target for iCCA therapy.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11229306/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141554209","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
CCDC88C, an O-GalNAc glycosylation substrate of GALNT6, drives breast cancer metastasis by promoting c-JUN-mediated CEMIP transcription. CCDC88C是GALNT6的O-GalNAc糖基化底物,它通过促进c-JUN介导的CEMIP转录推动乳腺癌转移。
IF 5.3 2区 医学 Q1 ONCOLOGY Pub Date : 2024-07-06 DOI: 10.1186/s12935-024-03413-2
Boya Deng, Siyang Zhang, Yingying Zhou, Ting Sun, Ying Zhu, Jing Fei, Ailin Li, Yuan Miao

Coiled-coil domain containing 88C (CCDC88C) is a component of non-canonical Wnt signaling, and its dysregulation causes colorectal cancer metastasis. Dysregulated expression of CCDC88C was observed in lymph node metastatic tumor tissues of breast cancer. However, the role of CCDC88C in breast cancer metastasis remains unclear. To address this, the stable BT549 and SKBR3 cell lines with CCDC88C overexpression or knockdown were developed. Loss/gain-of-function experiments suggested that CCDC88C drove breast cancer cell motility in vitro and lung and liver metastasis in vivo. We found that CCDC88C led to c-JUN-induced transcription activation. Overlapping genes were identified from the genes modulated by CCDC88C and c-JUN. CEMIP, one of these overlapping genes, has been confirmed to confer breast cancer metastasis. We found that CCDC88C regulated CEMIP mRNA levels via c-JUN and it exerted pro-metastatic capabilities in a CEMIP-dependent manner. Moreover, we identified the CCDC88C as a substrate of polypeptide N-acetylgalactosaminyltransferase 6 (GALNT6). GALNT6 was positively correlated with CCDC88C protein abundance in the normal breast and breast cancer tissues, indicating that GALNT6 might be associated with expression patterns of CCDC88C in breast cancer. Our data demonstrated that GALNT6 maintained CCDC88C stability by promoting its O-linked glycosylation, and the modification was critical for the pro-metastatic potential of CCDC88C. CCDC88C also could mediate the pro-metastatic potential of GALNT6 in breast cancer. Collectively, our findings uncover that CCDC88C may increase the risk of breast cancer metastasis and elucidate the underlying molecular mechanisms.

含盘旋卷曲结构域88C(CCDC88C)是非典型Wnt信号转导的一个组成部分,其失调会导致结直肠癌转移。在乳腺癌淋巴结转移肿瘤组织中也观察到了 CCDC88C 的表达失调。然而,CCDC88C 在乳腺癌转移中的作用仍不清楚。为了解决这个问题,研究人员开发了 CCDC88C 过表达或基因敲除的稳定 BT549 和 SKBR3 细胞系。功能缺失/功能增强实验表明,CCDC88C 在体外驱动乳腺癌细胞运动,在体内驱动肺和肝转移。我们发现 CCDC88C 会导致 c-JUN 诱导的转录激活。从受 CCDC88C 和 c-JUN 调节的基因中发现了重叠基因。CEMIP是这些重叠基因之一,已被证实可导致乳腺癌转移。我们发现,CCDC88C 通过 c-JUN 调节 CEMIP mRNA 水平,并以 CEMIP 依赖性方式发挥促转移能力。此外,我们还发现 CCDC88C 是多肽 N-乙酰半乳糖氨基转移酶 6(GALNT6)的底物。在正常乳腺和乳腺癌组织中,GALNT6 与 CCDC88C 蛋白丰度呈正相关,这表明 GALNT6 可能与 CCDC88C 在乳腺癌中的表达模式有关。我们的数据表明,GALNT6通过促进CCDC88C的O-连接糖基化来维持其稳定性,而这种修饰对CCDC88C的促转移潜能至关重要。CCDC88C 还能介导 GALNT6 在乳腺癌中的促转移潜能。总之,我们的研究结果揭示了CCDC88C可能会增加乳腺癌转移的风险,并阐明了其潜在的分子机制。
{"title":"CCDC88C, an O-GalNAc glycosylation substrate of GALNT6, drives breast cancer metastasis by promoting c-JUN-mediated CEMIP transcription.","authors":"Boya Deng, Siyang Zhang, Yingying Zhou, Ting Sun, Ying Zhu, Jing Fei, Ailin Li, Yuan Miao","doi":"10.1186/s12935-024-03413-2","DOIUrl":"10.1186/s12935-024-03413-2","url":null,"abstract":"<p><p>Coiled-coil domain containing 88C (CCDC88C) is a component of non-canonical Wnt signaling, and its dysregulation causes colorectal cancer metastasis. Dysregulated expression of CCDC88C was observed in lymph node metastatic tumor tissues of breast cancer. However, the role of CCDC88C in breast cancer metastasis remains unclear. To address this, the stable BT549 and SKBR3 cell lines with CCDC88C overexpression or knockdown were developed. Loss/gain-of-function experiments suggested that CCDC88C drove breast cancer cell motility in vitro and lung and liver metastasis in vivo. We found that CCDC88C led to c-JUN-induced transcription activation. Overlapping genes were identified from the genes modulated by CCDC88C and c-JUN. CEMIP, one of these overlapping genes, has been confirmed to confer breast cancer metastasis. We found that CCDC88C regulated CEMIP mRNA levels via c-JUN and it exerted pro-metastatic capabilities in a CEMIP-dependent manner. Moreover, we identified the CCDC88C as a substrate of polypeptide N-acetylgalactosaminyltransferase 6 (GALNT6). GALNT6 was positively correlated with CCDC88C protein abundance in the normal breast and breast cancer tissues, indicating that GALNT6 might be associated with expression patterns of CCDC88C in breast cancer. Our data demonstrated that GALNT6 maintained CCDC88C stability by promoting its O-linked glycosylation, and the modification was critical for the pro-metastatic potential of CCDC88C. CCDC88C also could mediate the pro-metastatic potential of GALNT6 in breast cancer. Collectively, our findings uncover that CCDC88C may increase the risk of breast cancer metastasis and elucidate the underlying molecular mechanisms.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":null,"pages":null},"PeriodicalIF":5.3,"publicationDate":"2024-07-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11227718/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141544577","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