首页 > 最新文献

Drug Resistance Updates最新文献

英文 中文
ABCB1-dependent collateral sensitivity of multidrug-resistant colorectal cancer cells to the survivin inhibitor MX106-4C 耐多药结肠直肠癌细胞对存活素抑制剂 MX106-4C 的 ABCB1 依赖性附带敏感性
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-02-06 DOI: 10.1016/j.drup.2024.101065
Zi-Ning Lei , Najah Albadari , Qiu-Xu Teng , Hadiar Rahman , Jing-Quan Wang , Zhongzhi Wu , Dejian Ma , Suresh V. Ambudkar , John N.D. Wurpel , Yihang Pan , Wei Li , Zhe-Sheng Chen

Aims

To investigate the collateral sensitivity (CS) of ABCB1-positive multidrug resistant (MDR) colorectal cancer cells to the survivin inhibitor MX106–4C and the mechanism.

Methods

Biochemical assays (MTT, ATPase, drug accumulation/efflux, Western blot, RT-qPCR, immunofluorescence, flow cytometry) and bioinformatic analyses (mRNA-sequencing, reversed-phase protein array) were performed to investigate the hypersensitivity of ABCB1 overexpressing colorectal cancer cells to MX106–4C and the mechanisms. Synergism assay, long-term selection, and 3D tumor spheroid test were used to evaluate the anti-cancer efficacy of MX106–4C.

Results

MX106–4C selectively killed ABCB1-positive colorectal cancer cells, which could be reversed by an ABCB1 inhibitor, knockout of ABCB1, or loss-of-function ABCB1 mutation, indicating an ABCB1 expression and function-dependent mechanism. MX106–4C's selective toxicity was associated with cell cycle arrest and apoptosis through ABCB1-dependent survivin inhibition and activation on caspases-3/7 as well as modulation on p21-CDK4/6-pRb pathway. MX106–4C had good selectivity against ABCB1-positive colorectal cancer cells and retained this in multicellular tumor spheroids. In addition, MX106–4C could exert a synergistic anti-cancer effect with doxorubicin or re-sensitize ABCB1-positive cancer cells to doxorubicin by reducing ABCB1 expression in the cell population via long-term exposure.

Conclusions

MX106–4C selectively kills ABCB1-positive MDR colorectal cancer cells via a novel ABCB1-dependent survivin inhibition mechanism, providing a clue for designing CS compound as an alternative strategy to overcome ABCB1-mediated colorectal cancer MDR.

目的 研究ABCB1阳性多药耐药(MDR)结直肠癌细胞对存活素抑制剂MX106-4C的附带敏感性(CS)及其机制。方法通过生化检测(MTT、ATPase、药物蓄积/外流、Western印迹、RT-qPCR、免疫荧光、流式细胞术)和生物信息学分析(mRNA测序、反相蛋白质阵列)研究ABCB1过表达结直肠癌细胞对MX106-4C的超敏性及其机制。结果MX106-4C能选择性杀伤ABCB1阳性结直肠癌细胞,ABCB1抑制剂、ABCB1基因敲除或ABCB1功能缺失突变均能逆转其杀伤作用,表明MX106-4C具有ABCB1表达和功能依赖性机制。MX106-4C的选择性毒性与细胞周期停滞和细胞凋亡有关,它是通过依赖于ABCB1的survivin抑制和caspases-3/7激活以及p21-CDK4/6-pRb通路调节实现的。MX106-4C对ABCB1阳性结直肠癌细胞具有良好的选择性,并在多细胞肿瘤球中保持了这种选择性。此外,MX106-4C 还能与多柔比星发挥协同抗癌作用,或通过长期暴露降低细胞群中 ABCB1 的表达,使 ABCB1 阳性癌细胞对多柔比星重新敏感。
{"title":"ABCB1-dependent collateral sensitivity of multidrug-resistant colorectal cancer cells to the survivin inhibitor MX106-4C","authors":"Zi-Ning Lei ,&nbsp;Najah Albadari ,&nbsp;Qiu-Xu Teng ,&nbsp;Hadiar Rahman ,&nbsp;Jing-Quan Wang ,&nbsp;Zhongzhi Wu ,&nbsp;Dejian Ma ,&nbsp;Suresh V. Ambudkar ,&nbsp;John N.D. Wurpel ,&nbsp;Yihang Pan ,&nbsp;Wei Li ,&nbsp;Zhe-Sheng Chen","doi":"10.1016/j.drup.2024.101065","DOIUrl":"https://doi.org/10.1016/j.drup.2024.101065","url":null,"abstract":"<div><h3>Aims</h3><p>To investigate the collateral sensitivity (CS) of ABCB1-positive multidrug resistant (MDR) colorectal cancer cells to the survivin inhibitor MX106–4C and the mechanism.</p></div><div><h3>Methods</h3><p>Biochemical assays (MTT, ATPase, drug accumulation/efflux, Western blot, RT-qPCR, immunofluorescence, flow cytometry) and bioinformatic analyses (mRNA-sequencing, reversed-phase protein array) were performed to investigate the hypersensitivity of ABCB1 overexpressing colorectal cancer cells to MX106–4C and the mechanisms. Synergism assay, long-term selection, and 3D tumor spheroid test were used to evaluate the anti-cancer efficacy of MX106–4C.</p></div><div><h3>Results</h3><p>MX106–4C selectively killed ABCB1-positive colorectal cancer cells, which could be reversed by an ABCB1 inhibitor, knockout of ABCB1, or loss-of-function ABCB1 mutation, indicating an ABCB1 expression and function-dependent mechanism. MX106–4C's selective toxicity was associated with cell cycle arrest and apoptosis through ABCB1-dependent survivin inhibition and activation on caspases-3/7 as well as modulation on p21-CDK4/6-pRb pathway. MX106–4C had good selectivity against ABCB1-positive colorectal cancer cells and retained this in multicellular tumor spheroids. In addition, MX106–4C could exert a synergistic anti-cancer effect with doxorubicin or re-sensitize ABCB1-positive cancer cells to doxorubicin by reducing ABCB1 expression in the cell population via long-term exposure.</p></div><div><h3>Conclusions</h3><p>MX106–4C selectively kills ABCB1-positive MDR colorectal cancer cells via a novel ABCB1-dependent survivin inhibition mechanism, providing a clue for designing CS compound as an alternative strategy to overcome ABCB1-mediated colorectal cancer MDR.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101065"},"PeriodicalIF":24.3,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139743913","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
Tumor-acquired somatic mutation affects conformation to abolish ABCG2-mediated drug resistance 肿瘤获得性体细胞突变会影响构象,从而消除 ABCG2 介导的耐药性。
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-02-06 DOI: 10.1016/j.drup.2024.101066
Tomoka Gose , Ali Rasouli , Sepehr Dehghani-Ghahnaviyeh , Po-Chao Wen , Yao Wang , John Lynch , Yu Fukuda , Talha Shafi , Robert C. Ford , Emad Tajkhorshid , John D. Schuetz

ABCG2 is an important ATP-binding cassette transporter impacting the absorption and distribution of over 200 chemical toxins and drugs. ABCG2 also reduces the cellular accumulation of diverse chemotherapeutic agents. Acquired somatic mutations in the phylogenetically conserved amino acids of ABCG2 might provide unique insights into its molecular mechanisms of transport. Here, we identify a tumor-derived somatic mutation (Q393K) that occurs in a highly conserved amino acid across mammalian species. This ABCG2 mutant seems incapable of providing ABCG2-mediated drug resistance. This was perplexing because it is localized properly and retained interaction with substrates and nucleotides. Using a conformationally sensitive antibody, we show that this mutant appears “locked” in a non-functional conformation. Structural modeling and molecular dynamics simulations based on ABCG2 cryo-EM structures suggested that the Q393K interacts with the E446 to create a strong salt bridge. The salt bridge is proposed to stabilize the inward-facing conformation, resulting in an impaired transporter that lacks the flexibility to readily change conformation, thereby disrupting the necessary communication between substrate binding and transport.

ABCG2 是一种重要的 ATP 结合盒转运体,影响着 200 多种化学毒素和药物的吸收和分布。ABCG2 还能减少多种化疗药物在细胞内的蓄积。ABCG2系统保守氨基酸的获得性体细胞突变可能会对其分子转运机制提供独特的见解。在这里,我们发现了一种来自肿瘤的体细胞突变(Q393K),这种突变发生在哺乳动物物种中高度保守的氨基酸上。这种ABCG2突变体似乎无法产生ABCG2介导的耐药性。这令人费解,因为它定位正确,并能保持与底物和核苷酸的相互作用。利用构象敏感抗体,我们发现该突变体似乎被 "锁定 "在一个无功能的构象中。基于 ABCG2 低温电子显微镜结构的结构建模和分子动力学模拟表明,Q393K 与 E446 相互作用,形成了一个强大的盐桥。盐桥被认为能稳定内向构象,导致转运体功能受损,缺乏随时改变构象的灵活性,从而破坏了底物结合与转运之间的必要沟通。
{"title":"Tumor-acquired somatic mutation affects conformation to abolish ABCG2-mediated drug resistance","authors":"Tomoka Gose ,&nbsp;Ali Rasouli ,&nbsp;Sepehr Dehghani-Ghahnaviyeh ,&nbsp;Po-Chao Wen ,&nbsp;Yao Wang ,&nbsp;John Lynch ,&nbsp;Yu Fukuda ,&nbsp;Talha Shafi ,&nbsp;Robert C. Ford ,&nbsp;Emad Tajkhorshid ,&nbsp;John D. Schuetz","doi":"10.1016/j.drup.2024.101066","DOIUrl":"10.1016/j.drup.2024.101066","url":null,"abstract":"<div><p>ABCG2 is an important ATP-binding cassette transporter impacting the absorption and distribution of over 200 chemical toxins and drugs. ABCG2 also reduces the cellular accumulation of diverse chemotherapeutic agents. Acquired somatic mutations in the phylogenetically conserved amino acids of ABCG2 might provide unique insights into its molecular mechanisms of transport. Here, we identify a tumor-derived somatic mutation (Q393K) that occurs in a highly conserved amino acid across mammalian species. This ABCG2 mutant seems incapable of providing ABCG2-mediated drug resistance. This was perplexing because it is localized properly and retained interaction with substrates and nucleotides. Using a conformationally sensitive antibody, we show that this mutant appears “locked” in a non-functional conformation. Structural modeling and molecular dynamics simulations based on ABCG2 cryo-EM structures suggested that the Q393K interacts with the E446 to create a strong salt bridge. The salt bridge is proposed to stabilize the inward-facing conformation, resulting in an impaired transporter that lacks the flexibility to readily change conformation, thereby disrupting the necessary communication between substrate binding and transport.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101066"},"PeriodicalIF":24.3,"publicationDate":"2024-02-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139934065","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
Fibroblast growth factor receptor 1 inhibition suppresses pancreatic cancer chemoresistance and chemotherapy-driven aggressiveness 抑制成纤维细胞生长因子受体 1 可抑制胰腺癌的化疗耐药性和化疗驱动的侵袭性
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-02-04 DOI: 10.1016/j.drup.2024.101064
Qingxiang Lin , Andrea Serratore , Jin Niu , Shichen Shen , Tista Roy Chaudhuri , Wen Wee Ma , Jun Qu , Eugene S. Kandel , Robert M. Straubinger

Aims

Pancreatic ductal adenocarcinoma (PDAC) is often intrinsically-resistant to standard-of-care chemotherapies such as gemcitabine. Acquired gemcitabine resistance (GemR) can arise from treatment of initially-sensitive tumors, and chemotherapy can increase tumor aggressiveness. We investigated the molecular mechanisms of chemoresistance and chemotherapy-driven tumor aggressiveness, which are understood incompletely.

Methods

Differential proteomic analysis was employed to investigate chemotherapy-driven chemoresistance drivers and responses of PDAC cells and patient-derived tumor xenografts (PDX) having different chemosensitivities. We also investigated the prognostic value of FGFR1 expression in the efficacy of selective pan-FGFR inhibitor (FGFRi)-gemcitabine combinations.

Results

Quantitative proteomic analysis of a highly-GemR cell line revealed fibroblast growth factor receptor 1 (FGFR1) as the highest-expressed receptor tyrosine kinase. FGFR1 knockdown or FGFRi co-treatment enhanced gemcitabine efficacy and decreased GemR marker expression, implicating FGFR1 in augmentation of GemR. FGFRi treatment reduced PDX tumor progression and prolonged survival significantly, even in highly-resistant tumors in which neither single-agent showed efficacy. Gemcitabine exacerbated aggressiveness of highly-GemR tumors, based upon proliferation and metastatic markers. Combining FGFRi with gemcitabine or gemcitabine+nab-paclitaxel reversed tumor aggressiveness and progression, and prolonged survival significantly. In multiple PDAC PDXs, FGFR1 expression correlated with intrinsic tumor gemcitabine sensitivity.

Conclusion

FGFR1 drives chemoresistance and tumor aggressiveness, which FGFRi can reverse.

目的 胰腺导管腺癌(PDAC)通常对吉西他滨等标准化疗药物具有内在耐药性。获得性吉西他滨耐药性(GemR)可能源于对最初敏感肿瘤的治疗,而化疗会增加肿瘤的侵袭性。我们研究了化疗耐药性和化疗驱动的肿瘤侵袭性的分子机制,目前对这些机制的了解尚不全面。方法采用差异蛋白质组学分析研究了不同化疗敏感性的PDAC细胞和患者衍生肿瘤异种移植物(PDX)的化疗驱动的化疗耐药性驱动因素和反应。我们还研究了FGFR1的表达在选择性泛FGFR抑制剂(FGFRi)-吉西他滨联合用药的疗效中的预后价值。FGFR1敲除或FGFRi联合治疗增强了吉西他滨的疗效并降低了GemR标记物的表达,这表明FGFR1参与了GemR的增强。FGFRi治疗可减少PDX肿瘤的进展并显著延长存活时间,即使是在单药均无疗效的高耐药性肿瘤中也是如此。根据增殖和转移标记物,吉西他滨会加剧高度吉西他滨肿瘤的侵袭性。FGFRi 与吉西他滨或吉西他滨+纳布-紫杉醇联用可逆转肿瘤的侵袭性和进展,并显著延长生存期。在多个 PDAC PDXs 中,FGFR1 的表达与肿瘤内在的吉西他滨敏感性相关。
{"title":"Fibroblast growth factor receptor 1 inhibition suppresses pancreatic cancer chemoresistance and chemotherapy-driven aggressiveness","authors":"Qingxiang Lin ,&nbsp;Andrea Serratore ,&nbsp;Jin Niu ,&nbsp;Shichen Shen ,&nbsp;Tista Roy Chaudhuri ,&nbsp;Wen Wee Ma ,&nbsp;Jun Qu ,&nbsp;Eugene S. Kandel ,&nbsp;Robert M. Straubinger","doi":"10.1016/j.drup.2024.101064","DOIUrl":"10.1016/j.drup.2024.101064","url":null,"abstract":"<div><h3>Aims</h3><p>Pancreatic ductal adenocarcinoma (PDAC) is often intrinsically-resistant to standard-of-care chemotherapies such as gemcitabine. Acquired gemcitabine resistance (GemR) can arise from treatment of initially-sensitive tumors, and chemotherapy can increase tumor aggressiveness. We investigated the molecular mechanisms of chemoresistance and chemotherapy-driven tumor aggressiveness, which are understood incompletely.</p></div><div><h3>Methods</h3><p>Differential proteomic analysis was employed to investigate chemotherapy-driven chemoresistance drivers and responses of PDAC cells and patient-derived tumor xenografts (PDX) having different chemosensitivities. We also investigated the prognostic value of FGFR1 expression in the efficacy of selective pan-FGFR inhibitor (FGFRi)-gemcitabine combinations.</p></div><div><h3>Results</h3><p>Quantitative proteomic analysis of a highly-GemR cell line revealed fibroblast growth factor receptor 1 (FGFR1) as the highest-expressed receptor tyrosine kinase. FGFR1 knockdown or FGFRi co-treatment enhanced gemcitabine efficacy and decreased GemR marker expression, implicating FGFR1 in augmentation of GemR. FGFRi treatment reduced PDX tumor progression and prolonged survival significantly, even in highly-resistant tumors in which neither single-agent showed efficacy. Gemcitabine exacerbated aggressiveness of highly-GemR tumors, based upon proliferation and metastatic markers. Combining FGFRi with gemcitabine or gemcitabine+nab-paclitaxel reversed tumor aggressiveness and progression, and prolonged survival significantly. In multiple PDAC PDXs, FGFR1 expression correlated with intrinsic tumor gemcitabine sensitivity.</p></div><div><h3>Conclusion</h3><p>FGFR1 drives chemoresistance and tumor aggressiveness, which FGFRi can reverse.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101064"},"PeriodicalIF":24.3,"publicationDate":"2024-02-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139878453","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
Methylation of GPRC5A promotes liver metastasis and docetaxel resistance through activating mTOR signaling pathway in triple negative breast cancer GPRC5A 的甲基化通过激活 mTOR 信号通路促进三阴性乳腺癌的肝转移和多西他赛耐药性
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-02-01 DOI: 10.1016/j.drup.2024.101063
Xueqi Ou , Yeru Tan , Jindong Xie , Jingping Yuan , Xinpei Deng , Ruonan Shao , Cailu Song , Xi Cao , Xiaoming Xie , Rongfang He , Yuehua Li , Hailin Tang

Aims

This study aims to explore the function and mechanism of G Protein-coupled receptor class C group 5 member A (GPRC5A) in docetaxel-resistance and liver metastasis of breast cancer.

Methods

Single-cell RNA transcriptomic analysis and bioinformatic analysis are used to screen relevant genes in breast cancer metastatic hepatic specimens. MeRIP, dual-luciferase analysis and bioinformation were used to detect m6A modulation. Mass spectrometry (MS), co-inmunoprecipitation (co-IP) and immunofluorescence colocalization were executed to explore the mechanism of GPRC5A in breast cancer cells.

Result

GPRC5A was upregulated in triple-negative breast cancer (TNBC) and was associated with a poor prognosis. In vitro and in vivo experiments demonstrated that knockdown of GPRC5A alleviated metastasis and resistance to docetaxel in TNBC. Overexpression of GPRC5A had the opposite effects. The m6A methylation of GPRC5A mRNA was modulated by METTL3 and YTHDF1, which facilitates its translation. GPRC5A inhibited the ubiquitination-dependent degradation of LAMTOR1, resulting in the recruitment of mTORC1 to lysosomes and activating the mTORC1/p70s6k signaling pathway.

Conclusion

METTL3/YTHDF1 axis up-regulates GPRC5A expression by m6A methylation. GPRC5A activates mTORC1/p70s6k signaling pathway by recruiting mTORC1 to lysosomes, consequently promotes docetaxel-resistance and liver metastasis.

目的本研究旨在探索G蛋白偶联受体C类5组A成员(GPRC5A)在多西他赛耐药和乳腺癌肝转移中的功能和机制。方法采用单细胞RNA转录组分析和生物信息学分析筛选乳腺癌肝转移标本中的相关基因。采用 MeRIP、双荧光素酶分析和生物信息学方法检测 m6A 调控。结果GPRC5A在三阴性乳腺癌(TNBC)中上调,并与不良预后相关。体外和体内实验表明,敲除 GPRC5A 可减轻 TNBC 的转移和对多西他赛的耐药性。而过表达 GPRC5A 则会产生相反的效果。GPRC5A mRNA 的 m6A 甲基化受 METTL3 和 YTHDF1 的调节,这有利于其翻译。GPRC5A 抑制了 LAMTOR1 的泛素依赖性降解,导致 mTORC1 被招募到溶酶体,激活了 mTORC1/p70s6k 信号通路。GPRC5A通过招募mTORC1至溶酶体激活mTORC1/p70s6k信号通路,从而促进多西他赛耐药和肝转移。
{"title":"Methylation of GPRC5A promotes liver metastasis and docetaxel resistance through activating mTOR signaling pathway in triple negative breast cancer","authors":"Xueqi Ou ,&nbsp;Yeru Tan ,&nbsp;Jindong Xie ,&nbsp;Jingping Yuan ,&nbsp;Xinpei Deng ,&nbsp;Ruonan Shao ,&nbsp;Cailu Song ,&nbsp;Xi Cao ,&nbsp;Xiaoming Xie ,&nbsp;Rongfang He ,&nbsp;Yuehua Li ,&nbsp;Hailin Tang","doi":"10.1016/j.drup.2024.101063","DOIUrl":"10.1016/j.drup.2024.101063","url":null,"abstract":"<div><h3>Aims</h3><p>This study aims to explore the function and mechanism of G Protein-coupled receptor class C group 5 member A (GPRC5A) in docetaxel-resistance and liver metastasis of breast cancer.</p></div><div><h3>Methods</h3><p>Single-cell RNA transcriptomic analysis and bioinformatic analysis are used to screen relevant genes in breast cancer metastatic hepatic specimens. MeRIP, dual-luciferase analysis and bioinformation were used to detect m6A modulation. Mass spectrometry (MS), co-inmunoprecipitation (co-IP) and immunofluorescence colocalization were executed to explore the mechanism of GPRC5A in breast cancer cells.</p></div><div><h3>Result</h3><p>GPRC5A was upregulated in triple-negative breast cancer (TNBC) and was associated with a poor prognosis. In vitro and in vivo experiments demonstrated that knockdown of GPRC5A alleviated metastasis and resistance to docetaxel in TNBC. Overexpression of GPRC5A had the opposite effects. The m6A methylation of GPRC5A mRNA was modulated by METTL3 and YTHDF1, which facilitates its translation. GPRC5A inhibited the ubiquitination-dependent degradation of LAMTOR1, resulting in the recruitment of mTORC1 to lysosomes and activating the mTORC1/p70s6k signaling pathway.</p></div><div><h3>Conclusion</h3><p>METTL3/YTHDF1 axis up-regulates GPRC5A expression by m6A methylation. GPRC5A activates mTORC1/p70s6k signaling pathway by recruiting mTORC1 to lysosomes, consequently promotes docetaxel-resistance and liver metastasis.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101063"},"PeriodicalIF":24.3,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1368764624000219/pdfft?md5=bf4c6ebbb0ee5c7db7592e6ce4fb4306&pid=1-s2.0-S1368764624000219-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139655960","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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 role of ABCC10/MRP7 in anti-cancer drug resistance and beyond ABCC10 / MRP7 在抗癌药物耐药性及其他方面的作用
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-01-29 DOI: 10.1016/j.drup.2024.101062
Da-Qian Chen , Yuhao Xie , Lu-Qi Cao , Joshua S. Fleishman , Yang Chen , Tiesong Wu , Dong-Hua Yang

Multidrug resistance protein 7 (MRP7), also known as ATP-binding cassette (ABC) transporter subfamily C10 (ABCC10), is an ABC transporter that was first identified in 2001. ABCC10/MRP7 is a 171 kDa protein located on the basolateral membrane of cells. ABCC10/MRP7 consists of three transmembrane domains and two nucleotide binding domains. It mediates multidrug resistance of tumor cells to a variety of anticancer drugs by increasing drug efflux and results in reducing intracellular drug accumulation. The transport substrates of ABCC10/MRP7 include antineoplastic drugs such as taxanes, vinca alkaloids, and epothilone B, as well as endobiotics such as leukotriene C4 (LTC4) and estradiol 17 β-D-glucuronide. A variety of ABCC10/MRP7 inhibitors, including cepharanthine, imatinib, erlotinib, tariquidar, and sildenafil, can reverse ABCC10/MRP7-mediated MDR. Additionally, the presence or absence of ABCC10/MRP7 is also closely related to renal tubular dysfunction, obesity, and other diseases. In this review, we discuss: 1) Structure and functions of ABCC10/MRP7; 2) Known substrates and inhibitors of ABCC10/MRP7 and their potential therapeutic applications in cancer; and 3) Role of ABCC10/MRP7 in non-cancerous diseases.

多药耐药性蛋白 7(MRP7)又称 ATP 结合盒(ABC)转运体 C10 亚家族(ABCC10),是一种 ABC 转运体,于 2001 年首次被发现。ABCC10/MRP7 是一种 171 kDa 的蛋白质,位于细胞的基底侧膜上。ABCC10/MRP7 由三个跨膜结构域和两个核苷酸结合结构域组成。它通过增加药物外流来介导肿瘤细胞对多种抗癌药物的多药耐药性,从而减少细胞内的药物积累。ABCC10/MRP7 的转运底物包括紫杉类、长春花生物碱和埃博霉素 B 等抗肿瘤药物,以及白三烯 C4(LTC4)和雌二醇 17 β-D-葡萄糖醛酸苷等内生药物。多种 ABCC10/MRP7 抑制剂,包括西酞普兰、伊马替尼、厄洛替尼、他利奎达和西地那非,都能逆转 ABCC10/MRP7 介导的 MDR。此外,ABCC10/MRP7的存在与否还与肾小管功能障碍、肥胖和其他疾病密切相关。在本综述中,我们将讨论1)ABCC10/MRP7 的结构和功能;2)ABCC10/MRP7 的已知底物和抑制剂及其在癌症中的潜在治疗应用;以及 3)ABCC10/MRP7 在非癌症疾病中的作用。
{"title":"The role of ABCC10/MRP7 in anti-cancer drug resistance and beyond","authors":"Da-Qian Chen ,&nbsp;Yuhao Xie ,&nbsp;Lu-Qi Cao ,&nbsp;Joshua S. Fleishman ,&nbsp;Yang Chen ,&nbsp;Tiesong Wu ,&nbsp;Dong-Hua Yang","doi":"10.1016/j.drup.2024.101062","DOIUrl":"10.1016/j.drup.2024.101062","url":null,"abstract":"<div><p>Multidrug resistance protein 7 (MRP7), also known as ATP-binding cassette (ABC) transporter subfamily C10 (ABCC10), is an ABC transporter that was first identified in 2001. ABCC10/MRP7 is a 171 kDa protein located on the basolateral membrane of cells. ABCC10/MRP7 consists of three transmembrane domains and two nucleotide binding domains. It mediates multidrug resistance of tumor cells to a variety of anticancer drugs by increasing drug efflux and results in reducing intracellular drug accumulation. The transport substrates of ABCC10/MRP7 include antineoplastic drugs such as taxanes, vinca alkaloids, and epothilone B, as well as endobiotics such as leukotriene C4 (LTC<sub>4</sub>) and estradiol 17 β-D-glucuronide. A variety of ABCC10/MRP7 inhibitors, including cepharanthine, imatinib, erlotinib, tariquidar, and sildenafil, can reverse ABCC10/MRP7-mediated MDR. Additionally, the presence or absence of ABCC10/MRP7 is also closely related to renal tubular dysfunction, obesity, and other diseases. In this review, we discuss: 1) Structure and functions of ABCC10/MRP7; 2) Known substrates and inhibitors of ABCC10/MRP7 and their potential therapeutic applications in cancer; and 3) Role of ABCC10/MRP7 in non-cancerous diseases.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101062"},"PeriodicalIF":24.3,"publicationDate":"2024-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139640930","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
Phage-mediated colistin resistance in Acinetobacter baumannii 噬菌体介导的鲍曼不动杆菌对可乐定的耐药性
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-01-28 DOI: 10.1016/j.drup.2024.101061
Massimiliano Lucidi , Francesco Imperi , Irene Artuso , Giulia Capecchi , Cinzia Spagnoli , Daniela Visaggio , Giordano Rampioni , Livia Leoni , Paolo Visca

Aims

Antimicrobial resistance is a global threat to human health, and Acinetobacter baumannii is a paradigmatic example of how rapidly bacteria become resistant to clinically relevant antimicrobials. The emergence of multidrug-resistant A. baumannii strains has forced the revival of colistin as a last-resort drug, suddenly leading to the emergence of colistin resistance. We investigated the genetic and molecular basis of colistin resistance in A. baumannii, and the mechanisms implicated in its regulation and dissemination.

Methods

Comparative genomic analysis was combined with genetic, biochemical, and phenotypic assays to characterize Φ19606, an A. baumannii temperate bacteriophage that carries a colistin resistance gene.

Results

Ф19606 was detected in 41% of 523 A. baumannii complete genomes and demonstrated to act as a mobile vehicle of the colistin resistance gene eptA1, encoding a functional lipid A phosphoethanolamine transferase. The eptA1 gene is coregulated with its chromosomal homolog pmrC via the PmrAB two-component system and confers colistin resistance when induced by low calcium and magnesium levels. Resistance selection assays showed that the eptA1-harbouring phage Ф19606 promotes the emergence of spontaneous colistin-resistant mutants.

Conclusions

Φ19606 is an unprecedented example of a self-transmissible phage vector implicated in the dissemination of colistin resistance.

目的抗菌药耐药性是对人类健康的全球性威胁,鲍曼不动杆菌是细菌对临床相关抗菌药迅速产生耐药性的典型例子。对多种药物耐药的鲍曼不动杆菌菌株的出现迫使人们重新将秋水仙碱作为最后的治疗药物,这突然导致了秋水仙碱耐药性的出现。我们研究了鲍曼不动杆菌耐受秋水仙碱的遗传和分子基础,以及其调控和传播机制。结果 Ф19606在523个鲍曼不动杆菌完整基因组中的41%被检测到,并被证明是鲍曼不动杆菌耐药基因eptA1的移动载体,eptA1编码功能性脂质A磷乙醇胺转移酶。eptA1 基因通过 PmrAB 双组分系统与其染色体上的同源基因 pmrC 核心配对,在低钙和低镁水平的诱导下产生对可乐定的抗性。抗性选择试验表明,eptA1-harbouring噬菌体Ф19606促进了自发性耐秋水仙素突变体的出现。
{"title":"Phage-mediated colistin resistance in Acinetobacter baumannii","authors":"Massimiliano Lucidi ,&nbsp;Francesco Imperi ,&nbsp;Irene Artuso ,&nbsp;Giulia Capecchi ,&nbsp;Cinzia Spagnoli ,&nbsp;Daniela Visaggio ,&nbsp;Giordano Rampioni ,&nbsp;Livia Leoni ,&nbsp;Paolo Visca","doi":"10.1016/j.drup.2024.101061","DOIUrl":"10.1016/j.drup.2024.101061","url":null,"abstract":"<div><h3><em>Aims</em></h3><p>Antimicrobial resistance is a global threat to human health, and <em>Acinetobacter baumannii</em> is a paradigmatic example of how rapidly bacteria become resistant to clinically relevant antimicrobials. The emergence of multidrug-resistant <em>A</em>. <em>baumannii</em> strains has forced the revival of colistin as a last-resort drug, suddenly leading to the emergence of colistin resistance. We investigated the genetic and molecular basis of colistin resistance in <em>A. baumannii</em>, and the mechanisms implicated in its regulation and dissemination.</p></div><div><h3><em>Methods</em></h3><p>Comparative genomic analysis was combined with genetic, biochemical, and phenotypic assays to characterize Φ19606, an <em>A</em>. <em>baumannii</em> temperate bacteriophage that carries a colistin resistance gene.</p></div><div><h3><em>Results</em></h3><p>Ф19606 was detected in 41% of 523 <em>A</em>. <em>baumannii</em> complete genomes and demonstrated to act as a mobile vehicle of the colistin resistance gene <em>eptA1</em>, encoding a functional lipid A phosphoethanolamine transferase<em>.</em> The <em>eptA1</em> gene is coregulated with its chromosomal homolog <em>pmrC</em> via the PmrAB two-component system and confers colistin resistance when induced by low calcium and magnesium levels. Resistance selection assays showed that the <em>eptA1</em>-harbouring phage Ф19606 promotes the emergence of spontaneous colistin-resistant mutants.</p></div><div><h3><em>Conclusions</em></h3><p>Φ19606 is an unprecedented example of a self-transmissible phage vector implicated in the dissemination of colistin resistance.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101061"},"PeriodicalIF":24.3,"publicationDate":"2024-01-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1368764624000190/pdfft?md5=e2e2dfa626e3a5985be906d1dbff90e9&pid=1-s2.0-S1368764624000190-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139568611","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Target-based drug design strategies to overcome resistance to antiviral agents: opportunities and challenges 克服抗病毒药物耐药性的靶向药物设计策略:机遇与挑战
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-01-26 DOI: 10.1016/j.drup.2024.101053
Shaoqing Du , Xueping Hu , Luis Menéndez-Arias , Peng Zhan , Xinyong Liu

Viral infections have a major impact in human health. Ongoing viral transmission and escalating selective pressure have the potential to favor the emergence of vaccine- and antiviral drug-resistant viruses. Target-based approaches for the design of antiviral drugs can play a pivotal role in combating drug-resistant challenges. Drug design computational tools facilitate the discovery of novel drugs. This review provides a comprehensive overview of current drug design strategies employed in the field of antiviral drug resistance, illustrated through the description of a series of successful applications. These strategies include technologies that enhance compound-target affinity while minimizing interactions with mutated binding pockets. Furthermore, emerging approaches such as virtual screening, targeted protein/RNA degradation, and resistance analysis during drug design have been harnessed to curtail the emergence of drug resistance. Additionally, host targeting antiviral drugs offer a promising avenue for circumventing viral mutation. The widespread adoption of these refined drug design strategies will effectively address the prevailing challenge posed by antiviral drug resistance.

病毒感染对人类健康有着重大影响。持续的病毒传播和不断升级的选择性压力有可能导致疫苗和抗病毒药物耐药病毒的出现。基于靶点的抗病毒药物设计方法可在应对耐药性挑战方面发挥关键作用。药物设计计算工具有助于发现新型药物。本综述全面概述了当前在抗病毒药物耐药性领域采用的药物设计策略,并通过描述一系列成功的应用加以说明。这些策略包括增强化合物与目标亲和力的技术,同时尽量减少与变异结合口袋的相互作用。此外,虚拟筛选、靶向蛋白质/核糖核酸降解和药物设计过程中的耐药性分析等新兴方法已被用于遏制耐药性的出现。此外,宿主靶向抗病毒药物为规避病毒变异提供了一条前景广阔的途径。广泛采用这些完善的药物设计策略将有效解决抗病毒药物耐药性带来的普遍挑战。
{"title":"Target-based drug design strategies to overcome resistance to antiviral agents: opportunities and challenges","authors":"Shaoqing Du ,&nbsp;Xueping Hu ,&nbsp;Luis Menéndez-Arias ,&nbsp;Peng Zhan ,&nbsp;Xinyong Liu","doi":"10.1016/j.drup.2024.101053","DOIUrl":"10.1016/j.drup.2024.101053","url":null,"abstract":"<div><p>Viral infections have a major impact in human health. Ongoing viral transmission and escalating selective pressure have the potential to favor the emergence of vaccine- and antiviral drug-resistant viruses. Target-based approaches for the design of antiviral drugs can play a pivotal role in combating drug-resistant challenges. Drug design computational tools facilitate the discovery of novel drugs. This review provides a comprehensive overview of current drug design strategies employed in the field of antiviral drug resistance, illustrated through the description of a series of successful applications. These strategies include technologies that enhance compound-target affinity while minimizing interactions with mutated binding pockets. Furthermore, emerging approaches such as virtual screening, targeted protein/RNA degradation, and resistance analysis during drug design have been harnessed to curtail the emergence of drug resistance. Additionally, host targeting antiviral drugs offer a promising avenue for circumventing viral mutation. The widespread adoption of these refined drug design strategies will effectively address the prevailing challenge posed by antiviral drug resistance.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101053"},"PeriodicalIF":24.3,"publicationDate":"2024-01-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139568490","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
Polyphenol nanocomplex modulates lactate metabolic reprogramming and elicits immune responses to enhance cancer therapeutic effect 多酚纳米复合物可调节乳酸代谢重编程并激发免疫反应,从而增强癌症治疗效果
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-01-20 DOI: 10.1016/j.drup.2024.101060
Zhan Zhang , Xinnan Li , Weiqiang Liu , Guanglei Chen , Jinchi Liu , Qingtian Ma , Pengjie Hou , Lu Liang , Caigang Liu

Cancer lactate metabolic reprogramming induces an elevated level of extracellular lactate and H+, leading to an acidic immunosuppressive tumor microenvironment (TEM). High lactic acid level may affect the metabolic programs of various cells that comprise an antitumor immune response, therefore, restricting immune-mediated tumor destruction, and leading to therapeutic resistance and unsatisfactory prognosis. Here, we report a metal-phenolic coordination-based nanocomplex loaded with a natural polyphenol galloflavin, which inhibits the function of lactate dehydrogenase, reducing the production of lactic acid, and alleviating the acidic immunosuppressive TME. Besides, the co-entrapped natural polyphenol carnosic acid and the synthetic PEG-Ce6 polyphenol derivative (serving as a photosensitizer) could induce immunogenic cancer cell death upon laser irradiation, which further activates immune system and promotes immune cell recruitment and infiltration in tumor tissues. We demonstrated that this nanocomplex-based combinational therapy could reshape the TME and elicit immune responses in a murine breast cancer model, which provides a promising strategy to enhance the therapeutic efficiency of drug-resistant breast cancer.

癌症乳酸代谢重编程诱导细胞外乳酸和H+水平升高,导致酸性免疫抑制性肿瘤微环境(TEM)。高乳酸水平可能会影响构成抗肿瘤免疫反应的各种细胞的代谢程序,从而限制免疫介导的肿瘤破坏,导致治疗抵抗和不理想的预后。在这里,我们报告了一种负载天然多酚五倍子黄素的金属酚配位型纳米复合物,它能抑制乳酸脱氢酶的功能,减少乳酸的产生,缓解酸性免疫抑制TME。此外,共包裹的天然多酚肉桂酸和合成的 PEG-Ce6 多酚衍生物(作为光敏剂)在激光照射下可诱导免疫原性癌细胞死亡,从而进一步激活免疫系统,促进免疫细胞在肿瘤组织中的募集和浸润。我们证明,这种基于纳米复合物的组合疗法可以重塑TME,并在小鼠乳腺癌模型中激发免疫反应,这为提高耐药乳腺癌的治疗效率提供了一种前景广阔的策略。
{"title":"Polyphenol nanocomplex modulates lactate metabolic reprogramming and elicits immune responses to enhance cancer therapeutic effect","authors":"Zhan Zhang ,&nbsp;Xinnan Li ,&nbsp;Weiqiang Liu ,&nbsp;Guanglei Chen ,&nbsp;Jinchi Liu ,&nbsp;Qingtian Ma ,&nbsp;Pengjie Hou ,&nbsp;Lu Liang ,&nbsp;Caigang Liu","doi":"10.1016/j.drup.2024.101060","DOIUrl":"10.1016/j.drup.2024.101060","url":null,"abstract":"<div><p>Cancer lactate metabolic reprogramming induces an elevated level of extracellular lactate and H<sup>+</sup>, leading to an acidic immunosuppressive tumor microenvironment (TEM). High lactic acid level may affect the metabolic programs of various cells that comprise an antitumor immune response, therefore, restricting immune-mediated tumor destruction, and leading to therapeutic resistance and unsatisfactory prognosis. Here, we report a metal-phenolic coordination-based nanocomplex loaded with a natural polyphenol galloflavin, which inhibits the function of lactate dehydrogenase, reducing the production of lactic acid, and alleviating the acidic immunosuppressive TME. Besides, the co-entrapped natural polyphenol carnosic acid and the synthetic PEG-Ce6 polyphenol derivative (serving as a photosensitizer) could induce immunogenic cancer cell death upon laser irradiation, which further activates immune system and promotes immune cell recruitment and infiltration in tumor tissues. We demonstrated that this nanocomplex-based combinational therapy could reshape the TME and elicit immune responses in a murine breast cancer model, which provides a promising strategy to enhance the therapeutic efficiency of drug-resistant breast cancer.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101060"},"PeriodicalIF":24.3,"publicationDate":"2024-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S1368764624000189/pdfft?md5=c1adb273fb999561d308f0725bebd41e&pid=1-s2.0-S1368764624000189-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139505985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Utilizing non-coding RNA-mediated regulation of ATP binding cassette (ABC) transporters to overcome multidrug resistance to cancer chemotherapy 利用非编码 RNA 介导的 ATP 结合盒 (ABC) 转运体调控克服癌症化疗的多药耐药性
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-01-20 DOI: 10.1016/j.drup.2024.101058
Kenneth K.W. To , Zoufang Huang , Hang Zhang , Charles R. Ashby Jr. , Liwu Fu

Multidrug resistance (MDR) is one of the primary factors that produces treatment failure in patients receiving cancer chemotherapy. MDR is a complex multifactorial phenomenon, characterized by a decrease or abrogation of the efficacy of a wide spectrum of anticancer drugs that are structurally and mechanistically distinct. The overexpression of the ATP-binding cassette (ABC) transporters, notably ABCG2 and ABCB1, are one of the primary mediators of MDR in cancer cells, which promotes the efflux of certain chemotherapeutic drugs from cancer cells, thereby decreasing or abolishing their therapeutic efficacy. A number of studies have suggested that non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), play a pivotal role in mediating the upregulation of ABC transporters in certain MDR cancer cells. This review will provide updated information about the induction of ABC transporters due to the aberrant regulation of ncRNAs in cancer cells. We will also discuss the measurement and biological profile of circulating ncRNAs in various body fluids as potential biomarkers for predicting the response of cancer patients to chemotherapy. Sequence variations, such as alternative polyadenylation of mRNA and single nucleotide polymorphism (SNPs) at miRNA target sites, which may indicate the interaction of miRNA-mediated gene regulation with genetic variations to modulate the MDR phenotype, will be reviewed. Finally, we will highlight novel strategies that could be used to modulate ncRNAs and circumvent ABC transporter-mediated MDR.

多药耐药性(MDR)是导致癌症化疗患者治疗失败的主要因素之一。多药耐药性是一种复杂的多因素现象,其特点是多种抗癌药物的疗效降低或减弱,这些药物在结构上和机理上各不相同。ATP结合盒(ABC)转运体,特别是ABCG2和ABCB1的过度表达是癌细胞MDR的主要介质之一,它能促进某些化疗药物从癌细胞中外流,从而降低或取消其疗效。许多研究表明,非编码 RNA(ncRNA),尤其是微 RNA(miRNA)、长非编码 RNA(lncRNA)和环状 RNA(circRNA),在介导某些 MDR 癌细胞中 ABC 转运体的上调方面起着关键作用。本综述将提供有关癌细胞中 ncRNAs 的异常调控诱导 ABC 转运体的最新信息。我们还将讨论各种体液中循环 ncRNA 的测量和生物特征,这些 ncRNA 是预测癌症患者对化疗反应的潜在生物标志物。我们还将回顾mRNA的替代多腺苷酸化和miRNA靶位点的单核苷酸多态性(SNPs)等序列变异,这些变异可能表明miRNA介导的基因调控与遗传变异相互作用,从而调节MDR表型。最后,我们将重点介绍可用于调节 ncRNA 和规避 ABC 转运体介导的 MDR 的新策略。
{"title":"Utilizing non-coding RNA-mediated regulation of ATP binding cassette (ABC) transporters to overcome multidrug resistance to cancer chemotherapy","authors":"Kenneth K.W. To ,&nbsp;Zoufang Huang ,&nbsp;Hang Zhang ,&nbsp;Charles R. Ashby Jr. ,&nbsp;Liwu Fu","doi":"10.1016/j.drup.2024.101058","DOIUrl":"10.1016/j.drup.2024.101058","url":null,"abstract":"<div><p><span>Multidrug resistance (MDR) is one of the primary factors that produces </span>treatment<span><span><span> failure in patients receiving </span>cancer chemotherapy<span><span>. MDR is a complex multifactorial phenomenon, characterized by a decrease or abrogation of the efficacy of a wide spectrum of anticancer drugs that are structurally and mechanistically distinct. The overexpression of the ATP-binding cassette (ABC) transporters, notably </span>ABCG2<span> and ABCB1, are one of the primary mediators of MDR in cancer cells<span>, which promotes the efflux of certain chemotherapeutic drugs from cancer cells, thereby decreasing or abolishing their therapeutic efficacy. A number of studies have suggested that non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and </span></span></span></span>circular RNAs<span> (circRNAs), play a pivotal role in mediating the upregulation of ABC transporters in certain MDR cancer cells. This review will provide updated information about the induction of ABC transporters due to the aberrant regulation of ncRNAs in cancer cells. We will also discuss the measurement and biological profile of circulating ncRNAs in various body fluids as potential biomarkers for predicting the response of cancer patients to chemotherapy. Sequence variations, such as alternative polyadenylation<span> of mRNA and single nucleotide polymorphism (SNPs) at miRNA target sites, which may indicate the interaction of miRNA-mediated gene regulation with genetic variations to modulate the MDR phenotype, will be reviewed. Finally, we will highlight novel strategies that could be used to modulate ncRNAs and circumvent ABC transporter-mediated MDR.</span></span></span></p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101058"},"PeriodicalIF":24.3,"publicationDate":"2024-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139505866","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
Drug resistance mechanisms in dopamine agonist-resistant prolactin pituitary neuroendocrine tumors and exploration for new drugs 多巴胺激动剂耐药型催乳素垂体神经内分泌肿瘤的耐药机制及新药探索
IF 24.3 1区 医学 Q1 PHARMACOLOGY & PHARMACY Pub Date : 2024-01-19 DOI: 10.1016/j.drup.2024.101056
Jianhua Cheng , Weiyan Xie , Yiyuan Chen , Yingxuan Sun , Lei Gong , Hongyun Wang , Chuzhong Li , Yazhuo Zhang

Background

The treatment of dopamine agonists (DA) resistant prolactinomas remains a formidable challenge, as the mechanism of resistance is still unclear, and there are currently no viable alternative drug therapies available. This study seeks to investigate the mechanism of DA resistance in prolactinomas and identify new potentially effective drugs.

Methods

To explore the mechanism of DA resistance in prolactinomas, this study conducted transcriptome sequencing analysis on 27 cases of DA-resistant prolactinomas and 10 cases of sensitive prolactinomas. In addition, single-cell sequencing analysis was performed on 3 cases of DA-resistant prolactinomas and 3 cases of sensitive prolactinomas. Furthermore, to screen for potential therapeutic drugs, the study successfully established an organoids model for DA-resistant prolactinomas and screened 180 small molecule compounds using 8 organoids. The efficacy of the identified drugs was verified through various assays, including CCK-8, colony formation, CTG, and flow cytometry, and their mechanisms of action were confirmed through WB and IHC. The effectiveness of the identified drugs was evaluated both in vitro and in vivo.

Results

The results of transcriptome sequencing and single-cell sequencing analyses showed that DA resistance in prolactinomas is associated with the upregulation of the Focal Adhesion (FA) signaling pathway. Additionally, immunohistochemical validation revealed that FAK and Paxillin were significantly upregulated in DA-resistant prolactinomas. Screening of 180 small molecule compounds using 8 organoids identified Genistein as a potentially effective drug for DA-resistant prolactinomas. Experimental validation demonstrated that Genistein inhibited the proliferation of pituitary tumor cell lines and organoids and promoted apoptosis in pituitary tumor cells. Moreover, both the cell sequencing results and WB validation results of the drug-treated cells indicated that Genistein exerts its anti-tumor effect by inhibiting the FA pathway. In vivo, experiments also showed that Genistein can inhibit subcutaneous tumor formation.

Conclusion

DA resistance in prolactinomas is associated with upregulation of the Focal Adhesion (FA) signaling pathway, and Genistein can exert its anti-tumor effect by inhibiting the expression of the FA pathway.

背景多巴胺受体激动剂(DA)耐药的泌乳素瘤的治疗仍然是一项艰巨的挑战,因为其耐药机制尚不清楚,而且目前还没有可行的替代药物疗法。为了探索泌乳素瘤的DA耐药机制,本研究对27例DA耐药的泌乳素瘤和10例敏感的泌乳素瘤进行了转录组测序分析。此外,还对3例DA耐药泌乳素瘤和3例敏感泌乳素瘤进行了单细胞测序分析。此外,为了筛选潜在的治疗药物,该研究成功建立了耐DA催乳素瘤的器官组织模型,并利用8个器官组织筛选了180种小分子化合物。通过CCK-8、菌落形成、CTG和流式细胞术等多种检测方法验证了所发现药物的疗效,并通过WB和IHC证实了这些药物的作用机制。结果转录组测序和单细胞测序分析结果显示,催乳素瘤的DA耐药性与病灶粘附(FA)信号通路的上调有关。此外,免疫组化验证显示,FAK 和 Paxillin 在 DA 抗性催乳素瘤中显著上调。利用 8 个器官组织对 180 种小分子化合物进行了筛选,发现 Genistein 是治疗 DA 抗性催乳素瘤的潜在有效药物。实验验证表明,Genistein 能抑制垂体瘤细胞系和器官组织的增殖,并促进垂体瘤细胞的凋亡。此外,药物处理细胞的细胞测序结果和 WB 验证结果均表明,染料木素是通过抑制 FA 通路来发挥抗肿瘤作用的。结论催乳素瘤的抗药性与Focal Adhesion(FA)信号通路的上调有关,而Genistein可以通过抑制FA通路的表达来发挥抗肿瘤作用。
{"title":"Drug resistance mechanisms in dopamine agonist-resistant prolactin pituitary neuroendocrine tumors and exploration for new drugs","authors":"Jianhua Cheng ,&nbsp;Weiyan Xie ,&nbsp;Yiyuan Chen ,&nbsp;Yingxuan Sun ,&nbsp;Lei Gong ,&nbsp;Hongyun Wang ,&nbsp;Chuzhong Li ,&nbsp;Yazhuo Zhang","doi":"10.1016/j.drup.2024.101056","DOIUrl":"10.1016/j.drup.2024.101056","url":null,"abstract":"<div><h3>Background</h3><p>The treatment<span><span> of dopamine agonists<span> (DA) resistant prolactinomas remains a formidable challenge, as the mechanism of resistance is still unclear, and there are currently no viable alternative drug therapies available. This study seeks to investigate the mechanism of DA resistance in prolactinomas and identify new potentially effective </span></span>drugs.</span></p></div><div><h3>Methods</h3><p><span>To explore the mechanism of DA resistance in prolactinomas, this study conducted transcriptome sequencing analysis on 27 cases of DA-resistant prolactinomas and 10 cases of sensitive prolactinomas. In addition, single-cell sequencing analysis was performed on 3 cases of DA-resistant prolactinomas and 3 cases of sensitive prolactinomas. Furthermore, to screen for potential therapeutic drugs, the study successfully established an </span>organoids<span> model for DA-resistant prolactinomas and screened 180 small molecule compounds using 8 organoids. The efficacy of the identified drugs was verified through various assays, including CCK-8, colony formation, CTG, and flow cytometry, and their mechanisms of action were confirmed through WB and IHC. The effectiveness of the identified drugs was evaluated both in vitro and in vivo.</span></p></div><div><h3>Results</h3><p>The results of transcriptome sequencing and single-cell sequencing analyses showed that DA resistance in prolactinomas is associated with the upregulation of the Focal Adhesion<span><span><span> (FA) signaling pathway. Additionally, immunohistochemical validation revealed that FAK and </span>Paxillin were significantly upregulated in DA-resistant prolactinomas. Screening of 180 small molecule compounds using 8 organoids identified </span>Genistein<span><span> as a potentially effective drug for DA-resistant prolactinomas. Experimental validation demonstrated that Genistein inhibited the proliferation of pituitary tumor cell lines and organoids and promoted </span>apoptosis in pituitary tumor cells. Moreover, both the cell sequencing results and WB validation results of the drug-treated cells indicated that Genistein exerts its anti-tumor effect by inhibiting the FA pathway. In vivo, experiments also showed that Genistein can inhibit subcutaneous tumor formation.</span></span></p></div><div><h3>Conclusion</h3><p>DA resistance in prolactinomas is associated with upregulation of the Focal Adhesion (FA) signaling pathway, and Genistein can exert its anti-tumor effect by inhibiting the expression of the FA pathway.</p></div>","PeriodicalId":51022,"journal":{"name":"Drug Resistance Updates","volume":"73 ","pages":"Article 101056"},"PeriodicalIF":24.3,"publicationDate":"2024-01-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139505822","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: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