首页 > 最新文献

Drug Resistance Updates最新文献

英文 中文
DEAF1 confers resistance to adriamycin-induced apoptosis and pyroptosis in multiple myeloma DEAF1对阿霉素诱导的多发性骨髓瘤细胞凋亡和焦亡具有抗性
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-16 DOI: 10.1016/j.drup.2025.101344
Zhendong Deng , Yongxin Wei , Shuang Liu , Lu Chen , Xuhui Wang , Zihao Liu , Lingling Liu , Yaohui Wang , Xinyu Lv , Shanliang Sun , Haiwen Ni , Chunyan Gu , Ye Yang

Aims

Transcriptional dysregulation by aberrant transcription factors (TFs) is a key driver of drug resistance. Resistance to adriamycin (ADR) frequently develops following first-line treatment for multiple myeloma (MM). This study aims to identify novel TFs associated with ADR resistance in MM and to elucidate their underlying mechanisms.

Methods

We employed a protein chip assay with FITC-labeled celastrol and identified the deformed epidermal autoregulatory factor 1 (DEAF1) as a potential target in MM. High-throughput sequencing was performed to identify DEAF1 downstream targets. Both in vivo and in vitro models were utilized to delineate the role of DEAF1 in MM cell proliferation and ADR resistance.

Results

High DEAF1 expression was associated with poor prognosis in MM patients, and was found to promote MM cell proliferation and induce ADR resistance. Mechanistically, DEAF1 directly binds to the RAD50 promoter via its SAND domain, upregulating RAD50 expression and consequently activating the ATM pathway. Furthermore, DEAF1 recruited AP-2-alpha (AP-2α) through its MYND domain, leading to the downregulation of tyrosine-protein kinase Fer (FER). This downregulation impaired FER-mediated phosphorylation of GSDME, which is known to enhance the cleavage efficiency of GSDME by caspase-3. Additionally, celastrol synergized with ADR to inhibit MM cell viability by disrupting the binding of DEAF1 to the promoters of its target genes.

Conclusions

Our findings demonstrate that DEAF1 attenuates ADR-induced apoptosis and pyroptosis in MM by enhancing DNA damage repair and suppressing GSDME cleavage via the FER/GSDME axis. This study provides a novel therapeutic target for the treatment of MM.
异常转录因子(TFs)介导的转录失调是耐药的关键驱动因素。多发性骨髓瘤(MM)的一线治疗后,阿霉素耐药性(ADR)经常出现。本研究旨在鉴定与MM耐ADR相关的新型tf,并阐明其潜在机制。方法采用fitc标记的celastrol蛋白芯片检测,鉴定变形表皮自调节因子1 (DEAF1)为MM的潜在靶点,并采用高通量测序方法鉴定DEAF1的下游靶点。利用体内和体外模型来描述DEAF1在MM细胞增殖和ADR抗性中的作用。结果耳聋1高表达与MM患者预后不良相关,并可促进MM细胞增殖,诱导ADR耐药。从机制上讲,DEAF1通过其SAND结构域直接与RAD50启动子结合,上调RAD50的表达,从而激活ATM通路。此外,DEAF1通过MYND结构域募集ap -2- α (AP-2α),导致酪氨酸蛋白激酶Fer (Fer)下调。这种下调破坏了ferr介导的GSDME磷酸化,而已知这可以提高caspase-3对GSDME的切割效率。此外,celastrol与ADR协同作用,通过破坏DEAF1与其靶基因启动子的结合来抑制MM细胞的活力。结论研究结果表明,DEAF1通过增强DNA损伤修复和抑制GSDME轴的裂解,从而减弱adr诱导的MM细胞凋亡和焦亡。本研究为MM的治疗提供了新的治疗靶点。
{"title":"DEAF1 confers resistance to adriamycin-induced apoptosis and pyroptosis in multiple myeloma","authors":"Zhendong Deng ,&nbsp;Yongxin Wei ,&nbsp;Shuang Liu ,&nbsp;Lu Chen ,&nbsp;Xuhui Wang ,&nbsp;Zihao Liu ,&nbsp;Lingling Liu ,&nbsp;Yaohui Wang ,&nbsp;Xinyu Lv ,&nbsp;Shanliang Sun ,&nbsp;Haiwen Ni ,&nbsp;Chunyan Gu ,&nbsp;Ye Yang","doi":"10.1016/j.drup.2025.101344","DOIUrl":"10.1016/j.drup.2025.101344","url":null,"abstract":"<div><h3>Aims</h3><div>Transcriptional dysregulation by aberrant transcription factors (TFs) is a key driver of drug resistance. Resistance to adriamycin (ADR) frequently develops following first-line treatment for multiple myeloma (MM). This study aims to identify novel TFs associated with ADR resistance in MM and to elucidate their underlying mechanisms.</div></div><div><h3>Methods</h3><div>We employed a protein chip assay with FITC-labeled celastrol and identified the deformed epidermal autoregulatory factor 1 (DEAF1) as a potential target in MM. High-throughput sequencing was performed to identify DEAF1 downstream targets. Both <em>in vivo</em> and <em>in vitro</em> models were utilized to delineate the role of DEAF1 in MM cell proliferation and ADR resistance.</div></div><div><h3>Results</h3><div>High DEAF1 expression was associated with poor prognosis in MM patients, and was found to promote MM cell proliferation and induce ADR resistance. Mechanistically, DEAF1 directly binds to the RAD50 promoter via its SAND domain, upregulating RAD50 expression and consequently activating the ATM pathway. Furthermore, DEAF1 recruited AP-2-alpha (AP-2α) through its MYND domain, leading to the downregulation of tyrosine-protein kinase Fer (FER). This downregulation impaired FER-mediated phosphorylation of GSDME, which is known to enhance the cleavage efficiency of GSDME by caspase-3. Additionally, celastrol synergized with ADR to inhibit MM cell viability by disrupting the binding of DEAF1 to the promoters of its target genes.</div></div><div><h3>Conclusions</h3><div>Our findings demonstrate that DEAF1 attenuates ADR-induced apoptosis and pyroptosis in MM by enhancing DNA damage repair and suppressing GSDME cleavage via the FER/GSDME axis. This study provides a novel therapeutic target for the treatment of MM.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101344"},"PeriodicalIF":21.7,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145786001","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Entinostat overcomes cisplatin resistance in bladder cancer by promoting H3K18la-mediated DHRS2 expression and nuclear translocation to suppress the AKR1C3-androgen axis 恩替诺他通过促进h3k18la介导的DHRS2表达和核易位抑制akr1c3 -雄激素轴来克服膀胱癌的顺铂耐药
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-09 DOI: 10.1016/j.drup.2025.101343
Guanghui Xu , Minghao Zheng , Zhigang Wu , Tianlei Xie , Yuqin Li , Ganlin Hu , Shuting Fang , Jing Zhang , Wenli Diao , Wei Zhao , Hongqian Guo , Junlong Zhuang
Epigenetic dysregulation is a significant factor contributing to cisplatin resistance in bladder cancer (BCa). Increasing studies indicated a synergistic effect of cisplatin and Entinostat, which is an FDA-approved histone deacetylases (HDAC) inhibitor, however, the underlying mechanisms of this effect remains unknown. Herein, the synergy of cisplatin and Entinostat was confirmed in BCa cells. Integrated RNA-seq and ATAC-seq analysis revealed that the combined regimen of cisplatin and Entinostat led to significant downregulation of platinum resistance and DNA damage repair-related pathways. We focused on the candidate gene dehydrogenase/reductase member 2 (DHRS2), and found that Entinostat counteracted cisplatin resistance via promoting histone H3K18 lactylation (H3K18la)-mediated DHRS2 upregulation and enhancing the nuclear translocation of DHRS2. DHRS2 downregulation promoted cisplatin resistance by upregulating aldo-keto reductase family 1 member C3 (AKR1C3), a key enzyme in androgen synthesis. Moreover, we validated a negative correlation between DHRS2 levels and AKR1C3 expression in clinical BCa samples. It was found that high DHRS2 and low AKR1C3 expression correlates with improved neoadjuvant chemotherapy (NAC) response. Furthermore, high DHRS2 predicts better survival specifically in male patients, indicating sex-specific androgen involvement. Overall, these findings elucidate the epigenetic mechanism underlying the cisplatin-sensitizing effect of Entinostat, and identifies the DHRS2–AKR1C3–androgen axis as a potential target, particularly for male patients.
表观遗传失调是膀胱癌(BCa)患者顺铂耐药的重要因素。越来越多的研究表明顺铂和恩替诺他具有协同作用,恩替诺他是fda批准的组蛋白去乙酰化酶(HDAC)抑制剂,然而,这种作用的潜在机制尚不清楚。本研究证实了顺铂和恩替诺他在BCa细胞中的协同作用。综合RNA-seq和ATAC-seq分析显示,顺铂和恩替诺他联合治疗方案导致铂耐药和DNA损伤修复相关通路的显著下调。我们重点研究候选基因脱氢酶/还原酶成员2 (DHRS2),发现恩替诺他通过促进组蛋白H3K18乳酸化(H3K18la)介导的DHRS2上调和增强DHRS2的核易位来对抗顺铂耐药。DHRS2下调通过上调醛酮还原酶家族1成员C3 (AKR1C3)促进顺铂耐药,AKR1C3是雄激素合成的关键酶。此外,我们验证了临床BCa样本中DHRS2水平与AKR1C3表达之间的负相关。研究发现,高DHRS2和低AKR1C3表达与改善新辅助化疗(NAC)反应相关。此外,高DHRS2特异性地预测男性患者更好的生存率,表明性别特异性雄激素参与。总的来说,这些发现阐明了恩替诺他顺铂致敏作用的表观遗传机制,并确定了dhrs2 - akr1c3雄激素轴是一个潜在的靶点,特别是对于男性患者。
{"title":"Entinostat overcomes cisplatin resistance in bladder cancer by promoting H3K18la-mediated DHRS2 expression and nuclear translocation to suppress the AKR1C3-androgen axis","authors":"Guanghui Xu ,&nbsp;Minghao Zheng ,&nbsp;Zhigang Wu ,&nbsp;Tianlei Xie ,&nbsp;Yuqin Li ,&nbsp;Ganlin Hu ,&nbsp;Shuting Fang ,&nbsp;Jing Zhang ,&nbsp;Wenli Diao ,&nbsp;Wei Zhao ,&nbsp;Hongqian Guo ,&nbsp;Junlong Zhuang","doi":"10.1016/j.drup.2025.101343","DOIUrl":"10.1016/j.drup.2025.101343","url":null,"abstract":"<div><div>Epigenetic dysregulation is a significant factor contributing to cisplatin resistance in bladder cancer (BCa). Increasing studies indicated a synergistic effect of cisplatin and Entinostat, which is an FDA-approved histone deacetylases (HDAC) inhibitor, however, the underlying mechanisms of this effect remains unknown. Herein, the synergy of cisplatin and Entinostat was confirmed in BCa cells. Integrated RNA-seq and ATAC-seq analysis revealed that the combined regimen of cisplatin and Entinostat led to significant downregulation of platinum resistance and DNA damage repair-related pathways. We focused on the candidate gene dehydrogenase/reductase member 2 (DHRS2), and found that Entinostat counteracted cisplatin resistance via promoting histone H3K18 lactylation (H3K18la)-mediated DHRS2 upregulation and enhancing the nuclear translocation of DHRS2. DHRS2 downregulation promoted cisplatin resistance by upregulating aldo-keto reductase family 1 member C3 (AKR1C3), a key enzyme in androgen synthesis. Moreover, we validated a negative correlation between DHRS2 levels and AKR1C3 expression in clinical BCa samples. It was found that high DHRS2 and low AKR1C3 expression correlates with improved neoadjuvant chemotherapy (NAC) response. Furthermore, high DHRS2 predicts better survival specifically in male patients, indicating sex-specific androgen involvement. Overall, these findings elucidate the epigenetic mechanism underlying the cisplatin-sensitizing effect of Entinostat, and identifies the DHRS2–AKR1C3–androgen axis as a potential target, particularly for male patients.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101343"},"PeriodicalIF":21.7,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145730716","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Hyperbaric oxygen targets RCN1 to modulate ER-mitochondria crosstalk and ameliorate sorafenib resistance in hepatocellular carcinoma 高压氧靶向RCN1调节er -线粒体串扰并改善肝细胞癌索拉非尼耐药
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-08 DOI: 10.1016/j.drup.2025.101342
Yuxi Lin , Feng Zhang , Weimin Guo , Jiaxin Guo , Xu Qiu , Yuelin Sun , Rong Li , Zihao Pan , Wentao Zhong , Min Yu , Jun Zheng , Jingwen Peng , Weifeng Hong
The emergence of resistance to tyrosine kinase inhibitors (TKIs) compromises their clinical benefits in patients with hepatocellular carcinoma (HCC), in part due to adaptive responses triggered by tumor hypoxia. In this study, we leverage hyperbaric oxygen (HBO) therapy as a sensitizing strategy in sorafenib-resistant HCC. We demonstrate that HBO significantly enhances the anti-tumor effect of sorafenib by stimulating calcium transfer from the endoplasmic reticulum (ER) to mitochondria, resulting in ER stress and mitochondrial dysfunction. Mechanistically, we show that hypoxia upregulates HNF4A, a transcriptional suppressor of RCN1, and HBO therapy effectively inhibits this hypoxia-driven HNF4A/RCN1 axis. Downregulation of RCN1, a calcium-binding protein overexpressed in sorafenib-resistant HCC, strengthens ER-mitochondria coupling. Subsequently, RCN1 suppression attenuates its interaction with IP3R1 through the EFh1/2 domain, facilitating IP3R1-GRP75 dissociation and the activation of mitochondrial calcium-uptake machinery. Using its EF-hand domains, RCN1 senses fluctuations in ER calcium concentration and accordingly employs a feedback mechanism to fine-tune its binding to IP3R1. In xenograft and spontaneous models, combined HBO-TKIs treatment delays tumor progression and modulates the HNF4A/RCN1 axis. Taken together, our findings elucidate a hitherto uncharacterized role of HBO in regulating ER-mitochondria calcium homeostasis and support its clinical application as an adjunctive therapy in TKI-resistant HCC.
酪氨酸激酶抑制剂(TKIs)耐药性的出现损害了其在肝细胞癌(HCC)患者中的临床益处,部分原因是肿瘤缺氧引发的适应性反应。在这项研究中,我们利用高压氧(HBO)治疗作为索拉非尼耐药HCC的增敏策略。我们证明,HBO通过刺激钙从内质网(ER)转移到线粒体,导致内质网应激和线粒体功能障碍,显著增强索拉非尼的抗肿瘤作用。在机制上,我们发现缺氧上调HNF4A (RCN1的转录抑制因子),而HBO治疗有效抑制了这种缺氧驱动的HNF4A/RCN1轴。RCN1是一种钙结合蛋白,在索拉非尼耐药HCC中过表达,下调RCN1可增强er -线粒体偶联。随后,RCN1抑制通过EFh1/2结构域减弱其与IP3R1的相互作用,促进IP3R1- grp75的解离和线粒体钙摄取机制的激活。RCN1利用其EF-hand结构域感知内质网钙浓度的波动,并相应地采用反馈机制微调其与IP3R1的结合。在异种移植和自发模型中,HBO-TKIs联合治疗延迟肿瘤进展并调节HNF4A/RCN1轴。综上所述,我们的研究结果阐明了HBO在调节内质网线粒体钙稳态中的作用,并支持其作为tki耐药HCC辅助治疗的临床应用。
{"title":"Hyperbaric oxygen targets RCN1 to modulate ER-mitochondria crosstalk and ameliorate sorafenib resistance in hepatocellular carcinoma","authors":"Yuxi Lin ,&nbsp;Feng Zhang ,&nbsp;Weimin Guo ,&nbsp;Jiaxin Guo ,&nbsp;Xu Qiu ,&nbsp;Yuelin Sun ,&nbsp;Rong Li ,&nbsp;Zihao Pan ,&nbsp;Wentao Zhong ,&nbsp;Min Yu ,&nbsp;Jun Zheng ,&nbsp;Jingwen Peng ,&nbsp;Weifeng Hong","doi":"10.1016/j.drup.2025.101342","DOIUrl":"10.1016/j.drup.2025.101342","url":null,"abstract":"<div><div>The emergence of resistance to tyrosine kinase inhibitors (TKIs) compromises their clinical benefits in patients with hepatocellular carcinoma (HCC), in part due to adaptive responses triggered by tumor hypoxia. In this study, we leverage hyperbaric oxygen (HBO) therapy as a sensitizing strategy in sorafenib-resistant HCC. We demonstrate that HBO significantly enhances the anti-tumor effect of sorafenib by stimulating calcium transfer from the endoplasmic reticulum (ER) to mitochondria, resulting in ER stress and mitochondrial dysfunction. Mechanistically, we show that hypoxia upregulates HNF4A, a transcriptional suppressor of RCN1, and HBO therapy effectively inhibits this hypoxia-driven HNF4A/RCN1 axis. Downregulation of RCN1, a calcium-binding protein overexpressed in sorafenib-resistant HCC, strengthens ER-mitochondria coupling. Subsequently, RCN1 suppression attenuates its interaction with IP3R1 through the EFh1/2 domain, facilitating IP3R1-GRP75 dissociation and the activation of mitochondrial calcium-uptake machinery. Using its EF-hand domains, RCN1 senses fluctuations in ER calcium concentration and accordingly employs a feedback mechanism to fine-tune its binding to IP3R1. In xenograft and spontaneous models, combined HBO-TKIs treatment delays tumor progression and modulates the HNF4A/RCN1 axis. Taken together, our findings elucidate a hitherto uncharacterized role of HBO in regulating ER-mitochondria calcium homeostasis and support its clinical application as an adjunctive therapy in TKI-resistant HCC.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101342"},"PeriodicalIF":21.7,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145731944","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
RCN2 facilitates esophageal squamous cellular carcinoma metastasis and cisplatin resistance through UBR5-mediated PPP2CA ubiquitination and degradation RCN2通过ubr5介导的PPP2CA泛素化和降解促进食管鳞癌转移和顺铂耐药
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-04 DOI: 10.1016/j.drup.2025.101339
Mengyuan Wu , Xu Huang , Miao Lin , Zhiyun Duan , Zitao Jian , Runze You , Peiyi Xie , Zhiwei Wu , Siyun Lin , Shaoyuan Zhang , Wenyi Xu , Heng Jiao , Han Tang , Lei Guo , Hao Wang , Weigang Guo , Lijie Tan

Aims

Metastatic progression and treatment resistance determine poor prognostic outcomes of patients with esophageal squamous cellular carcinoma (ESCC), highlighting the urgent need to understand the molecular mechanisms behind this. Reticulocalbin 2 (RCN2) is a calcium-binding protein localized in the endoplasmic reticulum lumen, which mediates tumor progression in various cancer types. However, the role of RCN2 in ESCC remains unexplored.

Methods

The influence of RCN2 on ESCC progression, metastasis, and cisplatin (CDDP) resistance was assessed both in vitro and in vivo. The downstream regulatory mechanism associated with RCN2 was screened through RNA-seq, TMT 10X mass spectrometry analysis, and LC-MS/MS analysis, which was further validated through Western blot, immunoprecipitation, immunofluorescence, GST pull-down assay, and rescue experiments.

Results

We observed high RCN2 expression in ESCC tumor tissues from patients with metastasis, which is correlated with a higher risk of metastasis and worse survival. PPP2CA, a catalytic subunit of protein phosphatase 2 A (PP2A), and ubiquitin protein ligase E3 component N-recognin 5 (UBR5) are determined as novel RCN2 functioning interactors. Mechanistically, RCN2 facilitates PPP2CA ubiquitination and degradation dependent on the HECT domain of UBR5, thereby activating the PI3K-AKT signaling pathway. Furthermore, the activated RCN2-PPP2CA-PI3K-AKT axis is validated in clinical specimens of ESCC. Finally, targeted suppression of RCN2 synergized with CDDP treatment to prevent tumor growth and metastasis in subcutaneous and lung metastasis models.

Conclusions

Overall, these findings identify RCN2 as a novel driver of ESCC metastasis and CDDP resistance. RCN2 could be a promising treatment target for ESCC.
转移进展和治疗耐药性决定了食管鳞状细胞癌(ESCC)患者预后不良的结果,强调了迫切需要了解其背后的分子机制。网状定位蛋白2 (Reticulocalbin 2, RCN2)是一种定位于内质网管腔的钙结合蛋白,在多种癌症类型中介导肿瘤进展。然而,RCN2在ESCC中的作用仍未被探索。方法体外和体内观察RCN2对ESCC进展、转移及顺铂耐药的影响。通过RNA-seq、TMT 10X质谱分析、LC-MS/MS分析筛选与RCN2相关的下游调控机制,并通过Western blot、免疫沉淀、免疫荧光、GST pull-down实验和救援实验进一步验证。结果我们在ESCC转移患者的肿瘤组织中观察到RCN2高表达,这与转移风险高和生存期差相关。蛋白磷酸酶2 a (PP2A)的催化亚基PPP2CA和泛素蛋白连接酶E3组分n -识别蛋白5 (UBR5)被确定为新的RCN2功能相互作用物。机制上,RCN2促进PPP2CA泛素化和降解依赖于UBR5的HECT结构域,从而激活PI3K-AKT信号通路。此外,激活的RCN2-PPP2CA-PI3K-AKT轴在ESCC临床标本中得到验证。最后,在皮下和肺转移模型中,靶向抑制RCN2与CDDP治疗协同抑制肿瘤生长和转移。总之,这些发现表明RCN2是ESCC转移和CDDP耐药的新驱动因素。RCN2可能是ESCC的一个有希望的治疗靶点。
{"title":"RCN2 facilitates esophageal squamous cellular carcinoma metastasis and cisplatin resistance through UBR5-mediated PPP2CA ubiquitination and degradation","authors":"Mengyuan Wu ,&nbsp;Xu Huang ,&nbsp;Miao Lin ,&nbsp;Zhiyun Duan ,&nbsp;Zitao Jian ,&nbsp;Runze You ,&nbsp;Peiyi Xie ,&nbsp;Zhiwei Wu ,&nbsp;Siyun Lin ,&nbsp;Shaoyuan Zhang ,&nbsp;Wenyi Xu ,&nbsp;Heng Jiao ,&nbsp;Han Tang ,&nbsp;Lei Guo ,&nbsp;Hao Wang ,&nbsp;Weigang Guo ,&nbsp;Lijie Tan","doi":"10.1016/j.drup.2025.101339","DOIUrl":"10.1016/j.drup.2025.101339","url":null,"abstract":"<div><h3>Aims</h3><div>Metastatic progression and treatment resistance determine poor prognostic outcomes of patients with esophageal squamous cellular carcinoma (ESCC), highlighting the urgent need to understand the molecular mechanisms behind this. Reticulocalbin 2 (RCN2) is a calcium-binding protein localized in the endoplasmic reticulum lumen, which mediates tumor progression in various cancer types. However, the role of RCN2 in ESCC remains unexplored.</div></div><div><h3>Methods</h3><div>The influence of RCN2 on ESCC progression, metastasis, and cisplatin (CDDP) resistance was assessed both <em>in vitro</em> and <em>in vivo</em>. The downstream regulatory mechanism associated with RCN2 was screened through RNA-seq, TMT 10X mass spectrometry analysis, and LC-MS/MS analysis, which was further validated through Western blot, immunoprecipitation, immunofluorescence, GST pull-down assay, and rescue experiments.</div></div><div><h3>Results</h3><div>We observed high RCN2 expression in ESCC tumor tissues from patients with metastasis, which is correlated with a higher risk of metastasis and worse survival. PPP2CA, a catalytic subunit of protein phosphatase 2 A (PP2A), and ubiquitin protein ligase E3 component N-recognin 5 (UBR5) are determined as novel RCN2 functioning interactors. Mechanistically, RCN2 facilitates PPP2CA ubiquitination and degradation dependent on the HECT domain of UBR5, thereby activating the PI3K-AKT signaling pathway. Furthermore, the activated RCN2-PPP2CA-PI3K-AKT axis is validated in clinical specimens of ESCC. Finally, targeted suppression of RCN2 synergized with CDDP treatment to prevent tumor growth and metastasis in subcutaneous and lung metastasis models.</div></div><div><h3>Conclusions</h3><div>Overall, these findings identify RCN2 as a novel driver of ESCC metastasis and CDDP resistance. RCN2 could be a promising treatment target for ESCC.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101339"},"PeriodicalIF":21.7,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145689546","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
UBL3 governs VEGFR inhibitor resistance by activating NOTCH signaling in renal cell carcinoma UBL3通过激活NOTCH信号调控肾细胞癌中VEGFR抑制剂的耐药性
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-12-02 DOI: 10.1016/j.drup.2025.101332
Diaoyi Tan , Yuzhong Ye , Daojia Miao , Chuanyi Zhao , Songming Wu , Jian Shi , Junkai Yang , Kanglin Fang , Feiyi Lu , Qingyang Lv , Jinshuo Gong , Hongmei Yang , Wen Xiao , Zhiyong Xiong , Xiaoping Zhang , Hailong Ruan

Background

Targeted therapy is the first-line treatment for patients with metastatic renal cell carcinoma (RCC), with vascular endothelial growth factor receptor inhibitors (VEGFRis) constituting the bulk of regimens used. Although the repertoire of VEGFRis for RCC now spans from sunitinib to cabozantinib, resistance to treatments has emerged as a common and prominent challenge. Thus, identifying novel therapeutic targets has become essential for enhancing the antitumor efficacy of current treatments and inhibiting RCC progression.

Method

To investigate the potential mechanisms underlying VEGFRi resistance in RCC, we performed a genome-wide CRISPR/Cas9 library screen under sunitinib and cabozantinib treatment and identified UBL3 as a key driver of VEGFRi resistance in RCC cells. The critical role of UBL3 in promoting VEGFRi resistance was validated using CCK8 assays, flow cytometry, TUNEL assays, and bioinformatics analyses. To elucidate the molecular mechanisms underlying UBL3, we utilized western blotting, RNA sequencing, chromatin immunoprecipitation, small extracellular vesicles (sEVs) isolation, and Astral-DIA proteomics. The contribution of UBL3 to VEGFRi resistance was further confirmed through comprehensive in vitro and in vivo experiments.

Results

UBL3 was confirmed to suppress apoptosis and promote VEGFRi resistance through NOTCH signaling activation. Further investigations highlighted the importance of NOTCH signaling in VEGFRi resistance in RCC via the NOTCH-PTEN-AKT and NOTCH-FOS pathways and revealed the mechanisms by which UBL3 activated NOTCH signaling. On the one hand, UBL3 formed complex with NOTCH2 and ADAM17 simultaneously, accelerating ADAM17-mediated cleavage of NOTCH2. On the other hand, UBL3-modified NOTCH2 was sorted into sEVs, which were taken up by recipient cells, activating NOTCH signaling and thereby transmitting VEGFRi resistance. Finally, lipid nanoparticle-mediated delivery of the CRISPR/Cas9 knockout system targeting UBL3 effectively restored the sensitivity of RCC tumors to VEGFRis.

Conclusion

This study emphasized the importance of UBL3 in VEGFRi resistance in RCC and proposed that UBL3 activated NOTCH signaling through two distinct pathways, thereby suppressing cancer apoptosis and promoting resistance to VEGFRis. These findings provided a solid scientific foundation and paved the way for the development of novel therapeutic strategies for patients with advanced RCC.
背景:靶向治疗是转移性肾细胞癌(RCC)患者的一线治疗方法,血管内皮生长因子受体抑制剂(VEGFRis)构成了使用的大部分方案。尽管VEGFRis治疗RCC的范围从舒尼替尼到卡博赞替尼,但对治疗的耐药性已经成为一个共同和突出的挑战。因此,确定新的治疗靶点对于增强当前治疗的抗肿瘤疗效和抑制RCC进展至关重要。为了研究VEGFRi在RCC中耐药的潜在机制,我们在舒尼替尼和卡博赞替尼治疗下进行了全基因组CRISPR/Cas9文库筛选,发现UBL3是RCC细胞中VEGFRi耐药的关键驱动因素。通过CCK8检测、流式细胞术、TUNEL检测和生物信息学分析验证了UBL3在促进VEGFRi耐药中的关键作用。为了阐明UBL3的分子机制,我们使用了western blotting、RNA测序、染色质免疫沉淀、小细胞外囊泡(sEVs)分离和Astral-DIA蛋白质组学。通过全面的体外和体内实验,进一步证实了UBL3对VEGFRi耐药的贡献。结果证实bl3通过NOTCH信号激活抑制细胞凋亡,促进VEGFRi耐药。进一步的研究强调了NOTCH信号通过NOTCH- pten - akt和NOTCH- fos通路在RCC中VEGFRi耐药中的重要性,并揭示了UBL3激活NOTCH信号的机制。一方面,UBL3与NOTCH2和ADAM17同时形成复合物,加速ADAM17介导的NOTCH2的裂解。另一方面,ubl3修饰的NOTCH2被分类成sev,被受体细胞占用,激活NOTCH信号,从而传递VEGFRi抗性。最后,脂质纳米颗粒介导的靶向UBL3的CRISPR/Cas9敲除系统有效地恢复了RCC肿瘤对VEGFRis的敏感性。本研究强调了UBL3在RCC中VEGFRi耐药中的重要性,并提出UBL3通过两种不同的途径激活NOTCH信号通路,从而抑制肿瘤凋亡,促进VEGFRi耐药。这些发现提供了坚实的科学基础,并为发展晚期RCC患者的新治疗策略铺平了道路。
{"title":"UBL3 governs VEGFR inhibitor resistance by activating NOTCH signaling in renal cell carcinoma","authors":"Diaoyi Tan ,&nbsp;Yuzhong Ye ,&nbsp;Daojia Miao ,&nbsp;Chuanyi Zhao ,&nbsp;Songming Wu ,&nbsp;Jian Shi ,&nbsp;Junkai Yang ,&nbsp;Kanglin Fang ,&nbsp;Feiyi Lu ,&nbsp;Qingyang Lv ,&nbsp;Jinshuo Gong ,&nbsp;Hongmei Yang ,&nbsp;Wen Xiao ,&nbsp;Zhiyong Xiong ,&nbsp;Xiaoping Zhang ,&nbsp;Hailong Ruan","doi":"10.1016/j.drup.2025.101332","DOIUrl":"10.1016/j.drup.2025.101332","url":null,"abstract":"<div><h3>Background</h3><div>Targeted therapy is the first-line treatment for patients with metastatic renal cell carcinoma (RCC), with vascular endothelial growth factor receptor inhibitors (VEGFRis) constituting the bulk of regimens used. Although the repertoire of VEGFRis for RCC now spans from sunitinib to cabozantinib, resistance to treatments has emerged as a common and prominent challenge. Thus, identifying novel therapeutic targets has become essential for enhancing the antitumor efficacy of current treatments and inhibiting RCC progression.</div></div><div><h3>Method</h3><div>To investigate the potential mechanisms underlying VEGFRi resistance in RCC, we performed a genome-wide CRISPR/Cas9 library screen under sunitinib and cabozantinib treatment and identified UBL3 as a key driver of VEGFRi resistance in RCC cells. The critical role of UBL3 in promoting VEGFRi resistance was validated using CCK8 assays, flow cytometry, TUNEL assays, and bioinformatics analyses. To elucidate the molecular mechanisms underlying UBL3, we utilized western blotting, RNA sequencing, chromatin immunoprecipitation, small extracellular vesicles (sEVs) isolation, and Astral-DIA proteomics. The contribution of UBL3 to VEGFRi resistance was further confirmed through comprehensive in vitro and in vivo experiments.</div></div><div><h3>Results</h3><div>UBL3 was confirmed to suppress apoptosis and promote VEGFRi resistance through NOTCH signaling activation. Further investigations highlighted the importance of NOTCH signaling in VEGFRi resistance in RCC via the NOTCH-PTEN-AKT and NOTCH-FOS pathways and revealed the mechanisms by which UBL3 activated NOTCH signaling. On the one hand, UBL3 formed complex with NOTCH2 and ADAM17 simultaneously, accelerating ADAM17-mediated cleavage of NOTCH2. On the other hand, UBL3-modified NOTCH2 was sorted into sEVs, which were taken up by recipient cells, activating NOTCH signaling and thereby transmitting VEGFRi resistance. Finally, lipid nanoparticle-mediated delivery of the CRISPR/Cas9 knockout system targeting UBL3 effectively restored the sensitivity of RCC tumors to VEGFRis.</div></div><div><h3>Conclusion</h3><div>This study emphasized the importance of UBL3 in VEGFRi resistance in RCC and proposed that UBL3 activated NOTCH signaling through two distinct pathways, thereby suppressing cancer apoptosis and promoting resistance to VEGFRis. These findings provided a solid scientific foundation and paved the way for the development of novel therapeutic strategies for patients with advanced RCC.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101332"},"PeriodicalIF":21.7,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145657171","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
The implied dysregulated RKIP-hypoxia axis in cancer and immune evasion: Clinical implications 癌症和免疫逃避中隐含的rkp -缺氧轴失调:临床意义
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-29 DOI: 10.1016/j.drup.2025.101328
Ryan McWhorter , Salem Chouaib , Benjamin Bonavida
The Raf kinase inhibitor protein (RKIP) functions as both a metastasis suppressor and immune enhancer, exerting its influence over several key oncogenic signaling pathways, including the MAPK, NF-κB, and PI3K pathways. Recent studies have highlighted a potential interplay between RKIP and hypoxia-inducible factors (HIFs), particularly in the hypoxic tumor microenvironment (TME). Hypoxia is known to reprogram cellular metabolism, enhance angiogenesis, and facilitate immune escape. Through analysis of cross-talk signaling pathways between RKIP and HIFs, we establish the presence of a dysregulated RKIP-hypoxia axis in cancer. Notably, many cancers simultaneously express low levels of RKIP and high levels of HIFs an expression pattern that strongly correlates with the emergence of immune evasion mechanisms. Herein, we report on the mechanisms by which this dysregulated axis mediates immune evasion. These include the molecular regulations of RKIP and HIFs expressions, and the low expression of RKIP and high expression of HIFs in several cancers. We report on the mechanisms underlying immune evasion by the RKIP-hypoxia axis by examining various factors intimately involved in immune evasion, such as the upregulation of PD-L1, matrix metalloproteinases (MMPs), anti-apoptotic molecules, CD47, and the enhanced frequencies of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), tumor-associated macrophage (TAM) polarization, and decreased antigen presentation. Thus, hypoxia-induced repression of RKIP establishes a feedforward loop that sustains immune evasion and tumor aggressiveness. Therapeutically, we propose that targeting the RKIP-hypoxia axis offers a new strategy to restore immune surveillance and counteract tumor progression. We present various means to target the inhibition of hypoxia as well as the induction of RKIP. Elucidating the molecular crosstalk between RKIP and hypoxic stress responses opens a new paradigm for strategies that enhance the efficacy of immunotherapies and overcome tumor resistance.
Raf激酶抑制剂蛋白(RKIP)作为转移抑制因子和免疫增强因子,对几种关键的致癌信号通路,包括MAPK、NF-κB和PI3K通路施加影响。最近的研究强调了RKIP与缺氧诱导因子(hfs)之间的潜在相互作用,特别是在缺氧肿瘤微环境(TME)中。众所周知,缺氧可以重新编程细胞代谢,增强血管生成,促进免疫逃逸。通过分析RKIP和hfs之间的串音信号通路,我们确定在癌症中存在一个失调的RKIP-缺氧轴。值得注意的是,许多癌症同时表达低水平的RKIP和高水平的hif,这种表达模式与免疫逃避机制的出现密切相关。在此,我们报告了这种失调轴介导免疫逃避的机制。其中包括RKIP和hif表达的分子调控,以及几种癌症中RKIP的低表达和hif的高表达。我们通过检查与免疫逃避密切相关的各种因素,如PD-L1、基质金属蛋白酶(MMPs)、抗凋亡分子、CD47的上调,以及调节性T细胞(Tregs)、髓源性抑制细胞(MDSCs)、肿瘤相关巨噬细胞(TAM)极化频率的增强,以及抗原呈递的减少,报道了rkip -缺氧轴免疫逃避的机制。因此,缺氧诱导的RKIP抑制建立了一个前馈循环,维持免疫逃避和肿瘤侵袭性。在治疗上,我们提出靶向rkip -缺氧轴提供了一种新的策略来恢复免疫监视和抑制肿瘤进展。我们提出了多种针对缺氧抑制和RKIP诱导的方法。阐明RKIP与低氧应激反应之间的分子串扰,为提高免疫治疗疗效和克服肿瘤耐药的策略开辟了新的范式。
{"title":"The implied dysregulated RKIP-hypoxia axis in cancer and immune evasion: Clinical implications","authors":"Ryan McWhorter ,&nbsp;Salem Chouaib ,&nbsp;Benjamin Bonavida","doi":"10.1016/j.drup.2025.101328","DOIUrl":"10.1016/j.drup.2025.101328","url":null,"abstract":"<div><div>The Raf kinase inhibitor protein (RKIP) functions as both a metastasis suppressor and immune enhancer, exerting its influence over several key oncogenic signaling pathways, including the MAPK, NF-κB, and PI3K pathways. Recent studies have highlighted a potential interplay between RKIP and hypoxia-inducible factors (HIFs), particularly in the hypoxic tumor microenvironment (TME). Hypoxia is known to reprogram cellular metabolism, enhance angiogenesis, and facilitate immune escape. Through analysis of cross-talk signaling pathways between RKIP and HIFs, we establish the presence of a dysregulated RKIP-hypoxia axis in cancer. Notably, many cancers simultaneously express low levels of RKIP and high levels of HIFs <img> an expression pattern that strongly correlates with the emergence of immune evasion mechanisms. Herein, we report on the mechanisms by which this dysregulated axis mediates immune evasion. These include the molecular regulations of RKIP and HIFs expressions, and the low expression of RKIP and high expression of HIFs in several cancers. We report on the mechanisms underlying immune evasion by the RKIP-hypoxia axis by examining various factors intimately involved in immune evasion, such as the upregulation of PD-L1, matrix metalloproteinases (MMPs), anti-apoptotic molecules, CD47, and the enhanced frequencies of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), tumor-associated macrophage (TAM) polarization, and decreased antigen presentation. Thus, hypoxia-induced repression of RKIP establishes a feedforward loop that sustains immune evasion and tumor aggressiveness. Therapeutically, we propose that targeting the RKIP-hypoxia axis offers a new strategy to restore immune surveillance and counteract tumor progression. We present various means to target the inhibition of hypoxia as well as the induction of RKIP. Elucidating the molecular crosstalk between RKIP and hypoxic stress responses opens a new paradigm for strategies that enhance the efficacy of immunotherapies and overcome tumor resistance.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101328"},"PeriodicalIF":21.7,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145613768","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Lars2-signaling mediates platinum resistance by accumulating cancer stem cell population and suppressing anti-tumor immunity lars2信号通过积累肿瘤干细胞群和抑制抗肿瘤免疫介导铂耐药
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-28 DOI: 10.1016/j.drup.2025.101330
Yuqing Wang , Min Deng , Haipeng Lei , Kai Miao , Xiaodong Shu , Jianjie Li , Dongyang Tang , Yangyang Feng , Sek Man Su , Ling Li , Yanjie Wang , Heng Sun , Fangyuan Shao , Tingting An , Xiaoling Li , Fanlin Zhou , Tingxiu Xiang , Xiaoling Xu , Chuxia Deng
Platinum-based chemotherapy remains a cornerstone of cancer treatment; however, its clinical efficacy is frequently compromised by acquired drug resistance. Our study elucidated a novel resistance mechanism mediated by LARS2 signaling in mammary tumors. Through comprehensive multi-omics analyses of cancer patients, mouse models, and functional validation, we demonstrated that platinum treatment upregulates LARS2 via a danger-triggered host response during resistant tumor progression, concomitant with increased chromatin accessibility. This signaling drives drug resistance through two key mechanisms: enrichment of cancer stem cells and promotion of TGF-β-mediated immunosuppression, as evidenced by M2 macrophage polarization and CD8+ T cell exhaustion. Importantly, we developed an effective therapeutic strategy combining carboplatin with LARS2 signaling pathway inhibition, which successfully reversed platinum resistance and restored PD-1 checkpoint blockade sensitivity in preclinical models. These findings not only advance our understanding of chemotherapy resistance, but also provide a translatable therapeutic framework for breast cancer and other platinum-treated malignancies.
以铂为基础的化疗仍然是癌症治疗的基石;然而,其临床疗效往往受到获得性耐药的影响。我们的研究阐明了LARS2信号在乳腺肿瘤中介导的一种新的耐药机制。通过对癌症患者、小鼠模型和功能验证的综合多组学分析,我们证明铂治疗在耐药肿瘤进展期间通过危险触发的宿主反应上调LARS2,同时增加染色质可及性。该信号通过两个关键机制驱动耐药:肿瘤干细胞的富集和TGF-β介导的免疫抑制的促进,M2巨噬细胞极化和CD8+ T细胞耗竭证明了这一点。重要的是,我们开发了一种有效的治疗策略,将卡铂与LARS2信号通路抑制相结合,在临床前模型中成功逆转了铂耐药并恢复了PD-1检查点阻断的敏感性。这些发现不仅促进了我们对化疗耐药的理解,而且为乳腺癌和其他铂治疗的恶性肿瘤提供了可翻译的治疗框架。
{"title":"Lars2-signaling mediates platinum resistance by accumulating cancer stem cell population and suppressing anti-tumor immunity","authors":"Yuqing Wang ,&nbsp;Min Deng ,&nbsp;Haipeng Lei ,&nbsp;Kai Miao ,&nbsp;Xiaodong Shu ,&nbsp;Jianjie Li ,&nbsp;Dongyang Tang ,&nbsp;Yangyang Feng ,&nbsp;Sek Man Su ,&nbsp;Ling Li ,&nbsp;Yanjie Wang ,&nbsp;Heng Sun ,&nbsp;Fangyuan Shao ,&nbsp;Tingting An ,&nbsp;Xiaoling Li ,&nbsp;Fanlin Zhou ,&nbsp;Tingxiu Xiang ,&nbsp;Xiaoling Xu ,&nbsp;Chuxia Deng","doi":"10.1016/j.drup.2025.101330","DOIUrl":"10.1016/j.drup.2025.101330","url":null,"abstract":"<div><div>Platinum-based chemotherapy remains a cornerstone of cancer treatment; however, its clinical efficacy is frequently compromised by acquired drug resistance. Our study elucidated a novel resistance mechanism mediated by LARS2 signaling in mammary tumors. Through comprehensive multi-omics analyses of cancer patients, mouse models, and functional validation, we demonstrated that platinum treatment upregulates LARS2 via a danger-triggered host response during resistant tumor progression, concomitant with increased chromatin accessibility. This signaling drives drug resistance through two key mechanisms: enrichment of cancer stem cells and promotion of TGF-β-mediated immunosuppression, as evidenced by M2 macrophage polarization and CD8<sup>+</sup> T cell exhaustion. Importantly, we developed an effective therapeutic strategy combining carboplatin with LARS2 signaling pathway inhibition, which successfully reversed platinum resistance and restored PD-1 checkpoint blockade sensitivity in preclinical models. These findings not only advance our understanding of chemotherapy resistance, but also provide a translatable therapeutic framework for breast cancer and other platinum-treated malignancies.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101330"},"PeriodicalIF":21.7,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145613764","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Glucocorticoid receptor activated by dexamethasone promotes the chemoresistance and stemness of lung cancer 地塞米松激活糖皮质激素受体促进肺癌的化疗耐药和干细胞
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-25 DOI: 10.1016/j.drup.2025.101331
Ting Yu , Dandan Peng , Xiao Liang , Wen Nie , Huaicheng Tan , Siyuan Chen , Huashan Shi , Yuquan Wei , Xiawei Wei

Aims

Glucocorticoids (GCs) such as dexamethasone are routinely used in patients to alleviate side effects of chemotherapeutic agents or symptoms caused by advanced cancer. However, growing evidences have found glucocorticoids-induced chemoresistance in solid tumors, while the potential effects and underlying mechanisms are still remained unclearly. This study aimed to reveal the underlying mechanism of glucocorticoids-induced chemoresistance in lung cancer.

Methods

Effects of dexamethasone on chemotherapy efficiency and stemness properties were tested both in vitro and in vivo assays. Underlying mechanism of dexamethasone was revealed by western blot, protein immunoprecipitation, molecular dynamics simulation, high-resolution mass spectrometry detection and RNA-sequencing. Prognostic value of glucocorticoid receptor (GR) activation in lung cancer patients was assessed through transcriptomic analyses of public datasets.

Results

Pre-treatment with dexamethasone significantly suppressed the apoptosis mediated by multiple chemotherapeutic agents in lung cancer cells. Pulmonary metastatic mouse models showed dexamethasone pre-treatment markedly reduced the anti-tumor efficiency of paclitaxel. Stemness-related properties of lung cancer were significantly improved after dexamethasone treatment, which manifested with enhanced self-renewal capability, improved chemoresistance, and increased tumor initiating potential in vivo. Moreover, we revealed the chemoresistance and stemness properties induced by dexamethasone were depended on GR-mediated nuclear translocation of β-catenin. The N-terminal domain (NTD) and activation function 2 (AF2) region of GR mediated the major contribution in the interaction with β-catenin. Analyses of clinical samples from TCGA-LUAD and GEO datasets demonstrated GR activation was associated with worse survival and less benefits from chemotherapy in lung cancer patients.

Conclusions

These results revealed dexamethasone could promote chemoresistance and stemness in lung cancer by inducing nuclear-translocation of GR/β-catenin complex. In the long run, more cautions are needed when glucocorticoids are prescribed to patients during chemotherapy.
目的糖皮质激素(GCs)如地塞米松通常用于减轻化疗药物的副作用或晚期癌症引起的症状。然而,越来越多的证据发现糖皮质激素在实体肿瘤中诱导化疗耐药,但其潜在影响和潜在机制尚不清楚。本研究旨在揭示糖皮质激素诱导肺癌化疗耐药的潜在机制。方法采用体外和体内试验,观察地塞米松对化疗疗效和干细胞特性的影响。通过western blot、蛋白免疫沉淀、分子动力学模拟、高分辨率质谱检测和rna测序等方法揭示地塞米松作用机制。通过对公共数据集的转录组学分析,评估肺癌患者糖皮质激素受体(GR)激活的预后价值。结果地塞米松预处理能显著抑制多种化疗药物介导的肺癌细胞凋亡。肺转移小鼠模型显示,地塞米松预处理明显降低紫杉醇的抗肿瘤效果。地塞米松治疗后,肺癌干细胞相关特性明显改善,表现为自我更新能力增强,化疗耐药改善,体内肿瘤启动电位增加。此外,我们发现地塞米松诱导的化学耐药和干性特性依赖于gr介导的β-连环蛋白核易位。GR的n端结构域(NTD)和激活功能2 (AF2)区域介导了与β-catenin相互作用的主要贡献。来自TCGA-LUAD和GEO数据集的临床样本分析表明,GR激活与肺癌患者更差的生存率和更少的化疗获益相关。结论地塞米松通过诱导GR/β-catenin复合物核易位,促进肺癌化疗耐药和干细胞的发生。从长远来看,在化疗期间给患者开糖皮质激素时需要更多的注意。
{"title":"Glucocorticoid receptor activated by dexamethasone promotes the chemoresistance and stemness of lung cancer","authors":"Ting Yu ,&nbsp;Dandan Peng ,&nbsp;Xiao Liang ,&nbsp;Wen Nie ,&nbsp;Huaicheng Tan ,&nbsp;Siyuan Chen ,&nbsp;Huashan Shi ,&nbsp;Yuquan Wei ,&nbsp;Xiawei Wei","doi":"10.1016/j.drup.2025.101331","DOIUrl":"10.1016/j.drup.2025.101331","url":null,"abstract":"<div><h3>Aims</h3><div>Glucocorticoids (GCs) such as dexamethasone are routinely used in patients to alleviate side effects of chemotherapeutic agents or symptoms caused by advanced cancer. However, growing evidences have found glucocorticoids-induced chemoresistance in solid tumors, while the potential effects and underlying mechanisms are still remained unclearly. This study aimed to reveal the underlying mechanism of glucocorticoids-induced chemoresistance in lung cancer.</div></div><div><h3>Methods</h3><div>Effects of dexamethasone on chemotherapy efficiency and stemness properties were tested both <em>in vitro</em> and <em>in vivo</em> assays. Underlying mechanism of dexamethasone was revealed by western blot, protein immunoprecipitation, molecular dynamics simulation, high-resolution mass spectrometry detection and RNA-sequencing. Prognostic value of glucocorticoid receptor (GR) activation in lung cancer patients was assessed through transcriptomic analyses of public datasets.</div></div><div><h3>Results</h3><div>Pre-treatment with dexamethasone significantly suppressed the apoptosis mediated by multiple chemotherapeutic agents in lung cancer cells. Pulmonary metastatic mouse models showed dexamethasone pre-treatment markedly reduced the anti-tumor efficiency of paclitaxel. Stemness-related properties of lung cancer were significantly improved after dexamethasone treatment, which manifested with enhanced self-renewal capability, improved chemoresistance, and increased tumor initiating potential <em>in vivo</em>. Moreover, we revealed the chemoresistance and stemness properties induced by dexamethasone were depended on GR-mediated nuclear translocation of β-catenin. The N-terminal domain (NTD) and activation function 2 (AF2) region of GR mediated the major contribution in the interaction with β-catenin. Analyses of clinical samples from TCGA-LUAD and GEO datasets demonstrated GR activation was associated with worse survival and less benefits from chemotherapy in lung cancer patients.</div></div><div><h3>Conclusions</h3><div>These results revealed dexamethasone could promote chemoresistance and stemness in lung cancer by inducing nuclear-translocation of GR/β-catenin complex. In the long run, more cautions are needed when glucocorticoids are prescribed to patients during chemotherapy.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"85 ","pages":"Article 101331"},"PeriodicalIF":21.7,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145598828","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Endothelial cells sense temozolomide resistance to facilitate monocyte-derived macrophage infiltration in glioblastoma 内皮细胞感知替莫唑胺耐药性,促进单核细胞来源的巨噬细胞浸润胶质母细胞瘤
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-22 DOI: 10.1016/j.drup.2025.101329
Wei Gao , Jianliang Huang , Kun Deng , Xiang Lin , Xinmiao Long , Xuetong Li , Meng Huang , Xiangyu Wang , Xiaoling She , Qing Liu , Minghua Wu

Aims

Glioblastoma (GBM), particularly mesenchymal and recurrent GBM, often develops resistance to temozolomide (TMZ) and is characterized by extensive infiltration of monocyte-derived macrophages (MDM), which contributes to treatment failure. However, the mechanisms through which TMZ-resistant GBM recruits MDM remain poorly understood. This study aims to investigate the molecular drivers of MDM infiltration in the context of TMZ resistance and to identify potential therapeutic targets to disrupt this process.

Methods

Patient-derived GBM organoid (GBO) was utilized as a model system. We performed molecular profiling to identify genes upregulated in TMZ-resistant recurrent GBO. Endothelial cells (ECs) cultures and preclinical GBM models were used to examine disruption of tight junctions and monocyte infiltration. Mechanistic studies employed genetic knockdown, pharmacological inhibition, and assays, including Chromatin immunoprecipitation-quantitative PCR, Western blot, and immunostaining, to validate pathway activity and protein interactions.

Results

COL6A1 (Collagen type VI alpha 1 chain) was significantly upregulated in TMZ-resistant recurrent GBO and associated with poor survival. COL6A1 is bound to ITGB1 (Integrin beta-1) on ECs, leading to disruption of tight junctions via UBD (Ubiquitin-like modifier D)-mediated degradation of claudin-5. Furthermore, COL6A1 activated the FAK/SRC/Hippo/YAP signaling axis, which promoted lactylation of the transcription factor IKZF1 (IKAROS family zinc finger 1) at lysine 255. Lactylated IKZF1 translocated to the nucleus and recruited the chromatin remodeler Chromodomain-helicase-DNA-binding protein 1 to enhance UBD transcription, thereby promoting endothelial barrier breakdown and monocyte infiltration. Treatment with lenalidomide (LEN), an IKZF1 inhibitor, restored claudin-5 expression, reduced MDM accumulation, and re-sensitized TMZ-resistant tumors to chemotherapy in preclinical models.

Conclusion

This study identifies a novel signaling cascade whereby TMZ-resistant GBM secretes COL6A1 to activate an IKZF1-UBD axis in ECs, disrupting blood vessel integrity and facilitating MDM infiltration. Our findings delineate the pivotal mechanism by which tumor cells engage ECs to drive MDM infiltration - a linchpin part of the positive-feedback loop that couples TMZ resistance to MDM influx. Targeting IKZF1 with LEN represents a promising strategy for restoring endothelial barrier function, reducing MDM infiltration, and enhancing chemosensitivity in GBM.
胶质母细胞瘤(GBM),特别是间充质和复发性GBM,经常对替莫唑胺(TMZ)产生耐药性,其特征是单核细胞源性巨噬细胞(MDM)的广泛浸润,这是治疗失败的原因之一。然而,对tmz抗性GBM招募MDM的机制仍然知之甚少。本研究旨在研究在TMZ耐药背景下MDM浸润的分子驱动因素,并确定破坏这一过程的潜在治疗靶点。方法采用患者源性GBM类器官(GBO)作为模型系统。我们进行了分子谱分析,以确定在耐tmz复发性GBO中上调的基因。内皮细胞(ECs)培养和临床前GBM模型用于检查紧密连接的破坏和单核细胞浸润。机制研究采用基因敲除、药理学抑制和分析,包括染色质免疫沉淀定量PCR、Western blot和免疫染色,以验证途径活性和蛋白质相互作用。结果col6a1 (VI型胶原α 1链)在tmz耐药复发性GBO中显著上调,并与较差的生存率相关。COL6A1与ECs上的ITGB1(整合素β -1)结合,通过UBD(泛素样修饰剂D)介导的claudin-5降解导致紧密连接中断。此外,COL6A1激活FAK/SRC/Hippo/YAP信号轴,促进转录因子IKZF1 (IKAROS家族锌指1)在赖氨酸255位点的乳酸化。乳酸化的IKZF1易位到细胞核,招募染色质重塑者染色体域解旋酶- dna结合蛋白1,增强UBD转录,从而促进内皮屏障的破坏和单核细胞的浸润。在临床前模型中,使用IKZF1抑制剂来那度胺(lenalidomide, LEN)治疗可以恢复claudin-5的表达,减少MDM积累,并使tmz耐药肿瘤对化疗重新敏感。本研究发现了一个新的信号级联,通过该信号级联,耐tmz的GBM分泌COL6A1激活ECs中的IKZF1-UBD轴,破坏血管完整性并促进MDM浸润。我们的研究结果描述了肿瘤细胞参与ECs驱动MDM浸润的关键机制,这是将TMZ抵抗MDM内流耦合在一起的正反馈回路的关键部分。用LEN靶向IKZF1是恢复内皮屏障功能、减少MDM浸润和增强GBM化疗敏感性的一种很有前景的策略。
{"title":"Endothelial cells sense temozolomide resistance to facilitate monocyte-derived macrophage infiltration in glioblastoma","authors":"Wei Gao ,&nbsp;Jianliang Huang ,&nbsp;Kun Deng ,&nbsp;Xiang Lin ,&nbsp;Xinmiao Long ,&nbsp;Xuetong Li ,&nbsp;Meng Huang ,&nbsp;Xiangyu Wang ,&nbsp;Xiaoling She ,&nbsp;Qing Liu ,&nbsp;Minghua Wu","doi":"10.1016/j.drup.2025.101329","DOIUrl":"10.1016/j.drup.2025.101329","url":null,"abstract":"<div><h3>Aims</h3><div>Glioblastoma (GBM), particularly mesenchymal and recurrent GBM, often develops resistance to temozolomide (TMZ) and is characterized by extensive infiltration of monocyte-derived macrophages (MDM), which contributes to treatment failure. However, the mechanisms through which TMZ-resistant GBM recruits MDM remain poorly understood. This study aims to investigate the molecular drivers of MDM infiltration in the context of TMZ resistance and to identify potential therapeutic targets to disrupt this process.</div></div><div><h3>Methods</h3><div>Patient-derived GBM organoid (GBO) was utilized as a model system. We performed molecular profiling to identify genes upregulated in TMZ-resistant recurrent GBO. Endothelial cells (ECs) cultures and preclinical GBM models were used to examine disruption of tight junctions and monocyte infiltration. Mechanistic studies employed genetic knockdown, pharmacological inhibition, and assays, including Chromatin immunoprecipitation-quantitative PCR, Western blot, and immunostaining, to validate pathway activity and protein interactions.</div></div><div><h3>Results</h3><div>COL6A1 (Collagen type VI alpha 1 chain) was significantly upregulated in TMZ-resistant recurrent GBO and associated with poor survival. COL6A1 is bound to ITGB1 (Integrin beta-1) on ECs, leading to disruption of tight junctions via UBD (Ubiquitin-like modifier D)-mediated degradation of claudin-5. Furthermore, COL6A1 activated the FAK/SRC/Hippo/YAP signaling axis, which promoted lactylation of the transcription factor IKZF1 (IKAROS family zinc finger 1) at lysine 255. Lactylated IKZF1 translocated to the nucleus and recruited the chromatin remodeler Chromodomain-helicase-DNA-binding protein 1 to enhance UBD transcription, thereby promoting endothelial barrier breakdown and monocyte infiltration. Treatment with lenalidomide (LEN), an IKZF1 inhibitor, restored claudin-5 expression, reduced MDM accumulation, and re-sensitized TMZ-resistant tumors to chemotherapy in preclinical models.</div></div><div><h3>Conclusion</h3><div>This study identifies a novel signaling cascade whereby TMZ-resistant GBM secretes COL6A1 to activate an IKZF1-UBD axis in ECs, disrupting blood vessel integrity and facilitating MDM infiltration. Our findings delineate the pivotal mechanism by which tumor cells engage ECs to drive MDM infiltration - a linchpin part of the positive-feedback loop that couples TMZ resistance to MDM influx. Targeting IKZF1 with LEN represents a promising strategy for restoring endothelial barrier function, reducing MDM infiltration, and enhancing chemosensitivity in GBM.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101329"},"PeriodicalIF":21.7,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145575463","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Integration of blaOXA-48 into a Col156 plasmid drove a carbapenem-resistant Escherichia coli ST131 outbreak in New Zealand: Global genomic evidence for the gene’s multilayered dissemination 将blaOXA-48整合到Col156质粒中,导致了新西兰耐碳青霉烯类大肠杆菌ST131的爆发:该基因多层次传播的全球基因组证据
IF 21.7 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-19 DOI: 10.1016/j.drup.2025.101327
Rhys T. White , Craig N. Thornley , Max Bloomfield , Kristin Dyet , Juliet Elvy , Hermes Perez , Allan Hardaker , Michael Harrington , Simon Jackson , Matthew Kelly , Loushy Mangalasseril , Annette Nesdale , Xiaoyun Ren , Jenny Szeto , Claire Underwood , David Winter , Rosemary Woodhouse , Zuyu Yang

Aims

To investigate the genetic diversity in OXA-48-producing Escherichia coli ST131 in a New Zealand community outbreak, and to characterize the mobile genetic elements carrying blaOXA-48, with emphasis on the gene’s global dissemination.

Methods

Forty outbreak isolates underwent short-read sequencing; 36 also underwent long-read sequencing. Bayesian phylogenetics reconstructed the emergence and spread of the outbreak. A pangenome graph of 543 Col156 plasmids and 806 global blaOXA-48-positive contigs were analyzed to assess structural diversity, mobility, and global distribution.

Results

The outbreak clone likely emerged circa 2017, following a single introduction into New Zealand after acquiring blaOXA-48 on a 7872 bp Col156 plasmid. It shares ancestry (circa 2009) with Southeast Asian E. coli ST131 genomes. Long-read sequencing and pangenome graph analyses identified a single IS1-mediated transposition of blaOXA-48 into a Col156 plasmid backbone, observed across species and continents. Globally, blaOXA-48 is present in diverse plasmid contexts and insertion sequence arrangements and is widely distributed among Enterobacterales.

Conclusions

This is the first high-resolution genomic reconstruction of a community-associated blaOXA-48 outbreak, identifying a compact Col156 plasmid as a key vector driving carbapenem resistance. Our findings demonstrate the value of complete genome assemblies and pangenome graph analyses in resolving the structural and evolutionary dynamics of antimicrobial resistance.
目的研究新西兰社区暴发中产生oxa -48的大肠杆菌ST131的遗传多样性,并表征携带blaOXA-48的移动遗传元件,重点研究该基因的全球传播。方法对40株暴发分离株进行短读测序;36个也进行了长读测序。贝叶斯系统发育重建了疫情的出现和传播。分析了543个Col156质粒和806个全球blaoxa -48阳性contigs的全基因组图,以评估结构多样性、流动性和全球分布。结果爆发克隆可能在2017年左右出现,在7872 bp Col156质粒上获得blaOXA-48后,被引入新西兰。它与东南亚大肠杆菌ST131基因组共享祖先(大约2009年)。长读测序和全基因组图分析发现,在不同物种和大洲中都观察到is1介导的blaOXA-48转位到Col156质粒主干。在全球范围内,blaOXA-48存在于不同的质粒背景和插入序列安排中,广泛分布于肠杆菌中。这是社区相关blaOXA-48暴发的第一个高分辨率基因组重建,确定了紧凑的Col156质粒是驱动碳青霉烯类耐药性的关键载体。我们的研究结果证明了全基因组组装和泛基因组图谱分析在解决抗菌素耐药性的结构和进化动力学方面的价值。
{"title":"Integration of blaOXA-48 into a Col156 plasmid drove a carbapenem-resistant Escherichia coli ST131 outbreak in New Zealand: Global genomic evidence for the gene’s multilayered dissemination","authors":"Rhys T. White ,&nbsp;Craig N. Thornley ,&nbsp;Max Bloomfield ,&nbsp;Kristin Dyet ,&nbsp;Juliet Elvy ,&nbsp;Hermes Perez ,&nbsp;Allan Hardaker ,&nbsp;Michael Harrington ,&nbsp;Simon Jackson ,&nbsp;Matthew Kelly ,&nbsp;Loushy Mangalasseril ,&nbsp;Annette Nesdale ,&nbsp;Xiaoyun Ren ,&nbsp;Jenny Szeto ,&nbsp;Claire Underwood ,&nbsp;David Winter ,&nbsp;Rosemary Woodhouse ,&nbsp;Zuyu Yang","doi":"10.1016/j.drup.2025.101327","DOIUrl":"10.1016/j.drup.2025.101327","url":null,"abstract":"<div><h3>Aims</h3><div>To investigate the genetic diversity in OXA-48-producing <em>Escherichia coli</em> ST131 in a New Zealand community outbreak, and to characterize the mobile genetic elements carrying <em>bla</em><sub>OXA-48</sub>, with emphasis on the gene’s global dissemination.</div></div><div><h3>Methods</h3><div>Forty outbreak isolates underwent short-read sequencing; 36 also underwent long-read sequencing. Bayesian phylogenetics reconstructed the emergence and spread of the outbreak. A pangenome graph of 543 Col156 plasmids and 806 global <em>bla</em><sub>OXA-48</sub>-positive contigs were analyzed to assess structural diversity, mobility, and global distribution.</div></div><div><h3>Results</h3><div>The outbreak clone likely emerged circa 2017, following a single introduction into New Zealand after acquiring <em>bla</em><sub>OXA-48</sub> on a 7872 bp Col156 plasmid. It shares ancestry (circa 2009) with Southeast Asian <em>E. coli</em> ST131 genomes. Long-read sequencing and pangenome graph analyses identified a single IS<em>1</em>-mediated transposition of <em>bla</em><sub>OXA-48</sub> into a Col156 plasmid backbone, observed across species and continents. Globally, <em>bla</em><sub>OXA-48</sub> is present in diverse plasmid contexts and insertion sequence arrangements and is widely distributed among Enterobacterales.</div></div><div><h3>Conclusions</h3><div>This is the first high-resolution genomic reconstruction of a community-associated <em>bla</em><sub>OXA-48</sub> outbreak, identifying a compact Col156 plasmid as a key vector driving carbapenem resistance. Our findings demonstrate the value of complete genome assemblies and pangenome graph analyses in resolving the structural and evolutionary dynamics of antimicrobial resistance.</div></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"84 ","pages":"Article 101327"},"PeriodicalIF":21.7,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145559712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
期刊
Drug Resistance Updates
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:604180095
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