首页 > 最新文献

BMC Pharmacology & Toxicology最新文献

英文 中文
Revealing the molecular mechanisms of levofloxacin-induced cognitive impairment and epilepsy: an integrated bioinformatics and molecular dynamics simulation. 揭示左氧氟沙星诱导认知障碍和癫痫的分子机制:综合生物信息学和分子动力学模拟。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-25 DOI: 10.1186/s40360-025-01025-8
Yixuan Zhang, Yimao Wu, Jing Du, Jingtao Chen, Haocheng Yu, Kaibo Yang, Fen Huang, Jianjun Xie, Jiangyun Peng
{"title":"Revealing the molecular mechanisms of levofloxacin-induced cognitive impairment and epilepsy: an integrated bioinformatics and molecular dynamics simulation.","authors":"Yixuan Zhang, Yimao Wu, Jing Du, Jingtao Chen, Haocheng Yu, Kaibo Yang, Fen Huang, Jianjun Xie, Jiangyun Peng","doi":"10.1186/s40360-025-01025-8","DOIUrl":"10.1186/s40360-025-01025-8","url":null,"abstract":"","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":"26 1","pages":"199"},"PeriodicalIF":2.7,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12648839/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145602223","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Selinexor, a selective inhibitor of nuclear export, shows anti-proliferative and anti-migratory effects on male germ cells in vitro. Selinexor是一种选择性核输出抑制剂,对体外雄性生殖细胞具有抗增殖和抗迁移作用。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-24 DOI: 10.1186/s40360-025-01034-7
Mustafa Öztatlıcı, Parmila Rahmat Zada, Rukiye Betül Çolaksel, Büşra Şen Halıcıoğlu, Hülya Öztatlıcı

Background: Selinexor (SLX), a selective inhibitor of nuclear export (SINE), has been shown to interfere with nuclear export mechanisms and to exert antitumor effects in a variety of cancer cell types. It is known to regulate multiple fundamental cellular processes, including the DNA damage response, cell proliferation, and stress signaling pathways. Nevertheless, its potential effects on reproductive cells remain inadequately characterized. The present study was aimed to investigate the cytotoxic, apoptotic, anti-proliferative and anti-migratory effects of SLX on GC1 (spermatogonia) and GC2 (spermatid) cell lines, alongside its influence on DNA damage and oxidative stress.

Methods: Cytotoxicity was assessed using the MTT assay. Cell proliferation capacity was evaluated via colony formation assay, while cell migration was analyzed using in vitro wound healing model. Apoptosis, oxidative stress, and DNA damage were investigated using immunocytochemical analyses of Cas-3, Bax, iNOS, ATM, and BRCA1 proteins. Additionally, Annexin V-FITC/PI staining was performed to detect the apoptotic cell population by flow cytometry.

Results: SLX treatment led to concentration- and time dependent cytotoxicity and colony formation assay revealed a marked reduction in proliferative capacity, in both cell lines. Wound healing analyses demonstrated that SLX effectively suppressed cell migration. Flow cytometry analysis showed that the live cell population decreased, whereas the late apoptotic cell population increased. Additionally, it was observed that Cas-3 and Bax immunoreactivities increased in the SLX groups compared to the control groups. Moreover, a significant increase in the immunoreactivity of ATM, BRCA1 and iNOS proteins, which are key indicators of DNA damage and oxidative stress, was observed.

Conclusion: The data suggest that SLX may decrease cell viability, induce apoptosis, inhibit cell migration and increase DNA damage and cellular stress in male germ cells. Given these effects, SLX should be carefully examined for its potential reproductive toxicity. Further studies are warranted to explore its long-term impact on male fertility.

背景:Selinexor (SLX)是一种选择性核输出抑制剂(SINE),已被证明可以干扰核输出机制,并在多种癌细胞类型中发挥抗肿瘤作用。众所周知,它调节多种基本的细胞过程,包括DNA损伤反应、细胞增殖和应激信号通路。然而,它对生殖细胞的潜在影响仍然没有充分的描述。本研究旨在探讨SLX对GC1(精原细胞)和GC2(精子)细胞系的细胞毒性、凋亡、抗增殖和抗迁移作用,以及对DNA损伤和氧化应激的影响。方法:采用MTT法测定细胞毒性。采用菌落形成法评估细胞增殖能力,采用体外创面愈合模型分析细胞迁移能力。通过免疫细胞化学分析cas3、Bax、iNOS、ATM和BRCA1蛋白,研究细胞凋亡、氧化应激和DNA损伤。流式细胞术采用Annexin V-FITC/PI染色检测凋亡细胞群。结果:SLX处理导致浓度和时间依赖的细胞毒性和集落形成试验显示,两种细胞系的增殖能力显著降低。伤口愈合分析表明,SLX有效抑制细胞迁移。流式细胞术分析显示活细胞数量减少,晚期凋亡细胞数量增加。此外,观察到与对照组相比,SLX组的cas3和Bax免疫反应活性增加。此外,我们还观察到,作为DNA损伤和氧化应激关键指标的ATM、BRCA1和iNOS蛋白的免疫反应性显著升高。结论:SLX可降低雄性生殖细胞活力,诱导细胞凋亡,抑制细胞迁移,增加DNA损伤和细胞应激。鉴于这些影响,应该仔细检查SLX的潜在生殖毒性。需要进一步研究其对男性生育能力的长期影响。
{"title":"Selinexor, a selective inhibitor of nuclear export, shows anti-proliferative and anti-migratory effects on male germ cells in vitro.","authors":"Mustafa Öztatlıcı, Parmila Rahmat Zada, Rukiye Betül Çolaksel, Büşra Şen Halıcıoğlu, Hülya Öztatlıcı","doi":"10.1186/s40360-025-01034-7","DOIUrl":"10.1186/s40360-025-01034-7","url":null,"abstract":"<p><strong>Background: </strong>Selinexor (SLX), a selective inhibitor of nuclear export (SINE), has been shown to interfere with nuclear export mechanisms and to exert antitumor effects in a variety of cancer cell types. It is known to regulate multiple fundamental cellular processes, including the DNA damage response, cell proliferation, and stress signaling pathways. Nevertheless, its potential effects on reproductive cells remain inadequately characterized. The present study was aimed to investigate the cytotoxic, apoptotic, anti-proliferative and anti-migratory effects of SLX on GC1 (spermatogonia) and GC2 (spermatid) cell lines, alongside its influence on DNA damage and oxidative stress.</p><p><strong>Methods: </strong>Cytotoxicity was assessed using the MTT assay. Cell proliferation capacity was evaluated via colony formation assay, while cell migration was analyzed using in vitro wound healing model. Apoptosis, oxidative stress, and DNA damage were investigated using immunocytochemical analyses of Cas-3, Bax, iNOS, ATM, and BRCA1 proteins. Additionally, Annexin V-FITC/PI staining was performed to detect the apoptotic cell population by flow cytometry.</p><p><strong>Results: </strong>SLX treatment led to concentration- and time dependent cytotoxicity and colony formation assay revealed a marked reduction in proliferative capacity, in both cell lines. Wound healing analyses demonstrated that SLX effectively suppressed cell migration. Flow cytometry analysis showed that the live cell population decreased, whereas the late apoptotic cell population increased. Additionally, it was observed that Cas-3 and Bax immunoreactivities increased in the SLX groups compared to the control groups. Moreover, a significant increase in the immunoreactivity of ATM, BRCA1 and iNOS proteins, which are key indicators of DNA damage and oxidative stress, was observed.</p><p><strong>Conclusion: </strong>The data suggest that SLX may decrease cell viability, induce apoptosis, inhibit cell migration and increase DNA damage and cellular stress in male germ cells. Given these effects, SLX should be carefully examined for its potential reproductive toxicity. Further studies are warranted to explore its long-term impact on male fertility.</p>","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":"26 1","pages":"196"},"PeriodicalIF":2.7,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12642124/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145595816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Cisplatin-induced toxicity in the hippocampus: a dose-dependent mechanism of damage. 顺铂诱导的海马毒性:剂量依赖性损伤机制。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-22 DOI: 10.1186/s40360-025-01050-7
Melek Altunkaya, Mehmet Burak Ateş, Ayşegül Bulut, Gülsüm Abuşoğlu, Bahadır Öztürk
{"title":"Cisplatin-induced toxicity in the hippocampus: a dose-dependent mechanism of damage.","authors":"Melek Altunkaya, Mehmet Burak Ateş, Ayşegül Bulut, Gülsüm Abuşoğlu, Bahadır Öztürk","doi":"10.1186/s40360-025-01050-7","DOIUrl":"https://doi.org/10.1186/s40360-025-01050-7","url":null,"abstract":"","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581869","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Development and validation of a UPLC-MS/MS method for the quantification of asciminib and its pharmacokinetic interaction and metabolic stability with shikonin. UPLC-MS/MS定量阿西米尼及其与紫草素的药动学相互作用和代谢稳定性的建立与验证。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-22 DOI: 10.1186/s40360-025-01049-0
Chenjian Zhou, Hailun Xia, Yingying Hu, Xiaohai Chen, Que Zou, Xuegu Xu, Zhe-Li Jiang
{"title":"Development and validation of a UPLC-MS/MS method for the quantification of asciminib and its pharmacokinetic interaction and metabolic stability with shikonin.","authors":"Chenjian Zhou, Hailun Xia, Yingying Hu, Xiaohai Chen, Que Zou, Xuegu Xu, Zhe-Li Jiang","doi":"10.1186/s40360-025-01049-0","DOIUrl":"https://doi.org/10.1186/s40360-025-01049-0","url":null,"abstract":"","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581806","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Insulin use in diabetes association with cognitive impairment and dementia incidence. 糖尿病患者胰岛素使用与认知障碍和痴呆发病率的关系。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-22 DOI: 10.1186/s40360-025-01042-7
Sajjad Hajihosseini, Dorsa Moharerzadeh Kurd, Fatemeh Nouroozi, Shayesteh Haghighi, Naeemeh Dini, Yeganeh Ghobadi, Negin Tehrani, Mehregan Shahrokhi, Mohaddeseh Belbasi, Haleh Alizadeh, Omid Salimi, Sepideh Hadimaleki, Danyal Yarahmadi, Niloofar Deravi
{"title":"Insulin use in diabetes association with cognitive impairment and dementia incidence.","authors":"Sajjad Hajihosseini, Dorsa Moharerzadeh Kurd, Fatemeh Nouroozi, Shayesteh Haghighi, Naeemeh Dini, Yeganeh Ghobadi, Negin Tehrani, Mehregan Shahrokhi, Mohaddeseh Belbasi, Haleh Alizadeh, Omid Salimi, Sepideh Hadimaleki, Danyal Yarahmadi, Niloofar Deravi","doi":"10.1186/s40360-025-01042-7","DOIUrl":"https://doi.org/10.1186/s40360-025-01042-7","url":null,"abstract":"","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581857","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Association between daily dose of dipeptidyl peptidase-4 inhibitors and change in glycated hemoglobin in patients with type 2 diabetes: interpretation of mixed-effects machine-learning models using electronic medical records. 二肽基肽酶-4抑制剂日剂量与2型糖尿病患者糖化血红蛋白变化之间的关系:利用电子病历解释混合效应机器学习模型
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-22 DOI: 10.1186/s40360-025-01055-2
Takashi Hayakawa, Hayato Akimoto, Takuya Nagashima, Kimino Minagawa, Yasuo Takahashi, Satoshi Asai
{"title":"Association between daily dose of dipeptidyl peptidase-4 inhibitors and change in glycated hemoglobin in patients with type 2 diabetes: interpretation of mixed-effects machine-learning models using electronic medical records.","authors":"Takashi Hayakawa, Hayato Akimoto, Takuya Nagashima, Kimino Minagawa, Yasuo Takahashi, Satoshi Asai","doi":"10.1186/s40360-025-01055-2","DOIUrl":"https://doi.org/10.1186/s40360-025-01055-2","url":null,"abstract":"","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581821","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Investigating the clinical efficacy, safety and molecular mechanism of sulforaphane in autism spectrum disorder: an integrated study combining meta-analysis, network pharmacology, and computational biology. 研究萝卜硫素治疗自闭症谱系障碍的临床疗效、安全性及分子机制:meta分析、网络药理学和计算生物学相结合的综合研究。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-22 DOI: 10.1186/s40360-025-01052-5
Junzi Long, Xingxing Liao, Zhiqing Tang, Kaiyue Han, Jiarou Chen, Xianna Wang, Jianjun Liu, Yan Zhang, Hao Zhang

Background: Sulforaphane, a natural antioxidant rich in cruciferous vegetables, has emerged as a promising dietary supplement for autism spectrum disorder (ASD). However, its therapeutic efficacy remains controversial, and the pharmacological mechanisms are not fully elucidated.

Methods: Eligible randomized controlled trials were retrieved from PubMed, Web of Science, Embase, and Cochrane Library databases. Review Manager 5.4 was used for meta-analysis and bias risk assessment. Network pharmacology, Mendelian randomization, GEO data analyses, molecular docking, and molecular dynamics simulation were employed to explore the mechanisms of sulforaphane in ASD.

Results: Six trials involving 333 participants were included in the meta-analysis. Pooled results demonstrated that both 4-5 weeks and 8-10 weeks of sulforaphane supplementation significantly decreased the scores on the Social Responsiveness Scale compared to placebo controls. No significant difference was observed in the incidence of adverse events. Network pharmacology identified 10 core targets of sulforaphane in ASD, including AKT1, EGFR, HSP90AA1, SRC, CASP3, STAT1, MAPK1, MMP9, MAPK8, and JAK2. These targets were implicated in the PI3K-Akt signaling pathway, MAPK signaling pathway, Chemokine signaling pathway, Chemical carcinogenesis - reactive oxygen species, TNF signaling pathway, Th17 cell differentiation, mTOR signaling pathway, and IL-17 signaling pathway. Mendelian randomization further revealed an inverse association between STAT1 levels and ASD risk. GEO transcriptomic data provided independent validation for the network pharmacology predictions. The binding energies between sulforaphane and the top 10 core targets are all ≤ -4.0 kcal/mol. Molecular dynamics simulations further validated the stable interaction between MMP-9 and sulforaphane.

Conclusion: Sulforaphane may serve as an efficacious and safe adjunctive therapy for ASD, mediated by its anti-oxidant and anti-inflammatory effects along with the modulation of autophagy.

Prospero registration number: CRD42025635045.

背景:萝卜硫素是十字花科蔬菜中富含的一种天然抗氧化剂,已成为治疗自闭症谱系障碍(ASD)的一种有前景的膳食补充剂。然而,其治疗效果仍有争议,药理机制尚未完全阐明。方法:从PubMed、Web of Science、Embase和Cochrane图书馆数据库中检索符合条件的随机对照试验。采用Review Manager 5.4进行meta分析和偏倚风险评估。采用网络药理学、孟德尔随机化、GEO数据分析、分子对接、分子动力学模拟等方法探讨萝卜硫素在ASD中的作用机制。结果:meta分析纳入了6项涉及333名受试者的试验。综合结果表明,与安慰剂对照组相比,4-5周和8-10周的萝卜硫素补充显著降低了社会反应量表的得分。两组不良事件发生率无显著差异。网络药理学鉴定出10个萝卜硫素在ASD中的核心靶点,包括AKT1、EGFR、HSP90AA1、SRC、CASP3、STAT1、MAPK1、MMP9、MAPK8和JAK2。这些靶点涉及PI3K-Akt信号通路、MAPK信号通路、趋化因子信号通路、化学致癌-活性氧、TNF信号通路、Th17细胞分化、mTOR信号通路和IL-17信号通路。孟德尔随机化进一步揭示了STAT1水平与ASD风险呈负相关。GEO转录组学数据为网络药理学预测提供了独立验证。萝卜硫素与前10位核心靶标的结合能均≤-4.0 kcal/mol。分子动力学模拟进一步验证了MMP-9与萝卜硫素之间稳定的相互作用。结论:萝卜硫素具有抗氧化、抗炎和调节自噬作用,可作为一种有效、安全的辅助治疗ASD的药物。普洛斯彼罗注册号:CRD42025635045。
{"title":"Investigating the clinical efficacy, safety and molecular mechanism of sulforaphane in autism spectrum disorder: an integrated study combining meta-analysis, network pharmacology, and computational biology.","authors":"Junzi Long, Xingxing Liao, Zhiqing Tang, Kaiyue Han, Jiarou Chen, Xianna Wang, Jianjun Liu, Yan Zhang, Hao Zhang","doi":"10.1186/s40360-025-01052-5","DOIUrl":"https://doi.org/10.1186/s40360-025-01052-5","url":null,"abstract":"<p><strong>Background: </strong>Sulforaphane, a natural antioxidant rich in cruciferous vegetables, has emerged as a promising dietary supplement for autism spectrum disorder (ASD). However, its therapeutic efficacy remains controversial, and the pharmacological mechanisms are not fully elucidated.</p><p><strong>Methods: </strong>Eligible randomized controlled trials were retrieved from PubMed, Web of Science, Embase, and Cochrane Library databases. Review Manager 5.4 was used for meta-analysis and bias risk assessment. Network pharmacology, Mendelian randomization, GEO data analyses, molecular docking, and molecular dynamics simulation were employed to explore the mechanisms of sulforaphane in ASD.</p><p><strong>Results: </strong>Six trials involving 333 participants were included in the meta-analysis. Pooled results demonstrated that both 4-5 weeks and 8-10 weeks of sulforaphane supplementation significantly decreased the scores on the Social Responsiveness Scale compared to placebo controls. No significant difference was observed in the incidence of adverse events. Network pharmacology identified 10 core targets of sulforaphane in ASD, including AKT1, EGFR, HSP90AA1, SRC, CASP3, STAT1, MAPK1, MMP9, MAPK8, and JAK2. These targets were implicated in the PI3K-Akt signaling pathway, MAPK signaling pathway, Chemokine signaling pathway, Chemical carcinogenesis - reactive oxygen species, TNF signaling pathway, Th17 cell differentiation, mTOR signaling pathway, and IL-17 signaling pathway. Mendelian randomization further revealed an inverse association between STAT1 levels and ASD risk. GEO transcriptomic data provided independent validation for the network pharmacology predictions. The binding energies between sulforaphane and the top 10 core targets are all ≤ -4.0 kcal/mol. Molecular dynamics simulations further validated the stable interaction between MMP-9 and sulforaphane.</p><p><strong>Conclusion: </strong>Sulforaphane may serve as an efficacious and safe adjunctive therapy for ASD, mediated by its anti-oxidant and anti-inflammatory effects along with the modulation of autophagy.</p><p><strong>Prospero registration number: </strong>CRD42025635045.</p>","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145581817","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Toxicity assessment of doxycycline-aided artificial intelligence-assisted drug design targeting candidate 16S rRNA methyltransferase gene. 靶向候选16S rRNA甲基转移酶基因的多西环素辅助人工智能辅助药物设计的毒性评估。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-21 DOI: 10.1186/s40360-025-00875-6
Hira Mubeen, Nagina Rafiq, Madiha Khan, Saima Jabeen, Muhammad Waseem Shoaib

Background: The misfunction of the protein 16SrRNA methyltransferase can result in Urinary tract infections (UTI), Gastrointestinal (GI) infections, sepsis, pneumonia, and wound infections; various tactics are used to lessen the fatal consequences. It confers resistance to aminoglycoside medications, which complicates the treatment of infections caused by these bacteria. Innovative methods are desperately needed to stop these diseases from spreading because there are no reliable medical therapies available.

Objectives: Herein, we aim to evaluate Doxycycline's Role in AI-Driven Drug Design and identification of effective inhibitors targeting the 16S rRNA methyltransferase gene. Additionally, to investigate the toxicological profiles of designed drug through AI approach for advancement in medical sciences.

Methodology: Methodology involves, selection of three effective de novo medicinal compounds that target the 16SrRNA methyltransferase protein for designing an AI driven drug. Multiple in silico tools were used for designing AI based drug includes: Expasy for protein annotation, ProtParam to calculate physiochemical parameters, SWISS-MODEL to estimate the 3D structure, and UniProt to generate the 16SrRNA methyltransferase protein sequence. An adequate foundation for the development and validation of AI-designed phytochemical medicines for infections is provided by quality assessment, binding site prediction, drug design with WADDAICA, toxicity screening, ADMET evaluation, and docking analysis with CB-dock.

Results: Comprehensive pharmacokinetic and toxicology analyses confirm that the AI-designed doxycycline exhibits a non-toxic character, with particularly high absorption through the blood-brain barrier. Furthermore, the AI-designed doxycycline docked complex demonstrates a strong docking affinity with the 16S rRNA methyltransferase protein, showing a binding energy of approximately - 7.6 kcal/mol, suggesting significant therapeutic potential.

Conclusion: Even though the in silico studies show efficacy and safety, still there is need of in vivo trials to investigate the hidden medical aspects. By addressing existing constraints, presenting a non-invasive approach to infections, and providing viable substitutes for traditional surgical procedures, this work considerably expands the knowledge about newer methods and also helps to understand deep insights of dug design mechanism for treatment.

背景:蛋白16SrRNA甲基转移酶功能障碍可导致尿路感染(UTI)、胃肠道感染(GI)、败血症、肺炎和伤口感染;为了减轻致命的后果,采取了各种策略。它使人对氨基糖苷类药物产生耐药性,从而使这些细菌引起的感染的治疗复杂化。由于没有可靠的医学治疗方法,迫切需要创新的方法来阻止这些疾病的传播。目的:在此,我们旨在评估强力霉素在人工智能驱动的药物设计中的作用,并鉴定针对16S rRNA甲基转移酶基因的有效抑制剂。此外,通过人工智能方法研究设计药物的毒理学特征,以促进医学科学的进步。方法:方法包括选择三种有效的靶向16SrRNA甲基转移酶蛋白的新药物化合物来设计人工智能驱动的药物。基于人工智能的药物设计使用了多种计算机工具,包括:Expasy用于蛋白质注释,ProtParam用于计算理化参数,SWISS-MODEL用于估计3D结构,UniProt用于生成16SrRNA甲基转移酶蛋白序列。通过质量评估、结合位点预测、WADDAICA药物设计、毒性筛选、ADMET评价、CB-dock对接分析,为ai设计的感染植物化学药物的开发和验证提供了充分的基础。结果:综合药代动力学和毒理学分析证实,ai设计的强力霉素具有无毒特性,通过血脑屏障的吸收率特别高。此外,ai设计的doxycycline对接复合物与16S rRNA甲基转移酶蛋白具有很强的对接亲和力,结合能约为- 7.6 kcal/mol,具有显著的治疗潜力。结论:尽管计算机研究显示了有效性和安全性,但仍需要进行体内试验来研究隐藏的医学方面。通过解决现有的限制,提出一种非侵入性的感染方法,并为传统的外科手术提供可行的替代品,这项工作大大扩展了对新方法的认识,也有助于深入了解治疗的挖设计机制。
{"title":"Toxicity assessment of doxycycline-aided artificial intelligence-assisted drug design targeting candidate 16S rRNA methyltransferase gene.","authors":"Hira Mubeen, Nagina Rafiq, Madiha Khan, Saima Jabeen, Muhammad Waseem Shoaib","doi":"10.1186/s40360-025-00875-6","DOIUrl":"10.1186/s40360-025-00875-6","url":null,"abstract":"<p><strong>Background: </strong>The misfunction of the protein 16SrRNA methyltransferase can result in Urinary tract infections (UTI), Gastrointestinal (GI) infections, sepsis, pneumonia, and wound infections; various tactics are used to lessen the fatal consequences. It confers resistance to aminoglycoside medications, which complicates the treatment of infections caused by these bacteria. Innovative methods are desperately needed to stop these diseases from spreading because there are no reliable medical therapies available.</p><p><strong>Objectives: </strong>Herein, we aim to evaluate Doxycycline's Role in AI-Driven Drug Design and identification of effective inhibitors targeting the 16S rRNA methyltransferase gene. Additionally, to investigate the toxicological profiles of designed drug through AI approach for advancement in medical sciences.</p><p><strong>Methodology: </strong>Methodology involves, selection of three effective de novo medicinal compounds that target the 16SrRNA methyltransferase protein for designing an AI driven drug. Multiple in silico tools were used for designing AI based drug includes: Expasy for protein annotation, ProtParam to calculate physiochemical parameters, SWISS-MODEL to estimate the 3D structure, and UniProt to generate the 16SrRNA methyltransferase protein sequence. An adequate foundation for the development and validation of AI-designed phytochemical medicines for infections is provided by quality assessment, binding site prediction, drug design with WADDAICA, toxicity screening, ADMET evaluation, and docking analysis with CB-dock.</p><p><strong>Results: </strong>Comprehensive pharmacokinetic and toxicology analyses confirm that the AI-designed doxycycline exhibits a non-toxic character, with particularly high absorption through the blood-brain barrier. Furthermore, the AI-designed doxycycline docked complex demonstrates a strong docking affinity with the 16S rRNA methyltransferase protein, showing a binding energy of approximately - 7.6 kcal/mol, suggesting significant therapeutic potential.</p><p><strong>Conclusion: </strong>Even though the in silico studies show efficacy and safety, still there is need of in vivo trials to investigate the hidden medical aspects. By addressing existing constraints, presenting a non-invasive approach to infections, and providing viable substitutes for traditional surgical procedures, this work considerably expands the knowledge about newer methods and also helps to understand deep insights of dug design mechanism for treatment.</p>","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":"26 1","pages":"195"},"PeriodicalIF":2.7,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12639713/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145572800","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Daflon attenuates cisplatin-induced cerebellar neurotoxicity, anxiety-like behavior, and motor dysfunction by downregulating TLR4/NF-kB signaling. 达芙莲通过下调TLR4/NF-kB信号通路,减轻顺铂诱导的小脑神经毒性、焦虑样行为和运动功能障碍。
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-21 DOI: 10.1186/s40360-025-01046-3
F B Fidelis, T M Akhigbe, A A Oladipo, P A Oyedokun, A S Lasisi-Sholola, O P Adepoju, O Ajao, O O Adeleye, O O Ogundipe, R E Akhigbe

Background: Although cisplatin is an effective chemotherapy, a major downside is its toxicity, including cerebellar neurotoxicity, which is mediated by the induction of inflammation and oxidative stress. On the other hand, daflon, a micronized purified flavonoid fraction, suppresses inflammation and oxidative stress. However, the effect of daflon on cisplatin-induced cerebellar neurotoxicity has not been documented.

Aim: The present study evaluated the effect of daflon in cisplatin-induced cerebellar toxicity. In addition, the role of TLR4/NF-kB signaling was explored.

Materials and methods: Twenty male Wistar rats were acclimatized for 2 weeks and then randomized into 4 equal groups: control, daflon-treated, cisplatin-treated, and cisplatin+daflon-treated.

Results: Daflon significantly improved cisplatin-induced distortions in cerebellar histology, evidenced by increased thickness in the molecular and intergranular layers, increased Purkinje cells, and reduced pyknotic neurons. Also, daflon attenuated cisplatin-induced rise in malondialdehyde and cisplatin-driven decline in glutathione, superoxide dismutase, and catalase activities. Furthermore, daflon ameliorated cisplatin-induced rise in myeloperoxidase activity and tumour-necrosis factor α, interleukin-1β 1β, and interleukin-6 levels. Additionally, daflon suppressed cisplatin-induced upregulation of toll-like receptor-4, nuclear factor-kappa B, cyclo-oxygenase-2, prostaglandin E2, and caspase-3 activity in the cerebellar tissue.

Conclusion: In conclusion, daflon confers neuroprotection against cisplatin-induced cerebellar neurotoxicity through the suppression of TLR4/NF-kB-mediated oxidative-inflammatory and apoptotic injury.

背景:虽然顺铂是一种有效的化疗药物,但其主要的缺点是其毒性,包括小脑神经毒性,这是由诱导炎症和氧化应激介导的。另一方面,蒲公英,一个微粉纯化类黄酮的部分,抑制炎症和氧化应激。然而,达芙莲对顺铂诱导的小脑神经毒性的影响尚未被证实。目的:评价蒲公英对顺铂所致小脑毒性的影响。此外,我们还探讨了TLR4/NF-kB信号的作用。材料与方法:将20只雄性Wistar大鼠驯化2周后,随机分为对照组、达芙蓉组、顺铂组、顺铂+达芙蓉组。结果:Daflon显著改善了顺铂诱导的小脑组织学扭曲,表现为分子层和粒间层厚度增加,浦肯野细胞增加,收缩神经元减少。此外,达芬还能减弱顺铂引起的丙二醛升高和顺铂引起的谷胱甘肽、超氧化物歧化酶和过氧化氢酶活性下降。此外,蒲公英改善了顺铂诱导的髓过氧化物酶活性、肿瘤坏死因子α、白细胞介素-1β 1β和白细胞介素-6水平的升高。此外,蒲公英抑制顺铂诱导的小脑组织toll样受体-4、核因子- κ B、环氧化酶-2、前列腺素E2和caspase-3活性的上调。结论:蒲公英通过抑制TLR4/ nf - kb介导的氧化性炎症和凋亡损伤,对顺铂诱导的小脑神经毒性具有神经保护作用。
{"title":"Daflon attenuates cisplatin-induced cerebellar neurotoxicity, anxiety-like behavior, and motor dysfunction by downregulating TLR4/NF-kB signaling.","authors":"F B Fidelis, T M Akhigbe, A A Oladipo, P A Oyedokun, A S Lasisi-Sholola, O P Adepoju, O Ajao, O O Adeleye, O O Ogundipe, R E Akhigbe","doi":"10.1186/s40360-025-01046-3","DOIUrl":"https://doi.org/10.1186/s40360-025-01046-3","url":null,"abstract":"<p><strong>Background: </strong>Although cisplatin is an effective chemotherapy, a major downside is its toxicity, including cerebellar neurotoxicity, which is mediated by the induction of inflammation and oxidative stress. On the other hand, daflon, a micronized purified flavonoid fraction, suppresses inflammation and oxidative stress. However, the effect of daflon on cisplatin-induced cerebellar neurotoxicity has not been documented.</p><p><strong>Aim: </strong>The present study evaluated the effect of daflon in cisplatin-induced cerebellar toxicity. In addition, the role of TLR4/NF-kB signaling was explored.</p><p><strong>Materials and methods: </strong>Twenty male Wistar rats were acclimatized for 2 weeks and then randomized into 4 equal groups: control, daflon-treated, cisplatin-treated, and cisplatin+daflon-treated.</p><p><strong>Results: </strong>Daflon significantly improved cisplatin-induced distortions in cerebellar histology, evidenced by increased thickness in the molecular and intergranular layers, increased Purkinje cells, and reduced pyknotic neurons. Also, daflon attenuated cisplatin-induced rise in malondialdehyde and cisplatin-driven decline in glutathione, superoxide dismutase, and catalase activities. Furthermore, daflon ameliorated cisplatin-induced rise in myeloperoxidase activity and tumour-necrosis factor α, interleukin-1β 1β, and interleukin-6 levels. Additionally, daflon suppressed cisplatin-induced upregulation of toll-like receptor-4, nuclear factor-kappa B, cyclo-oxygenase-2, prostaglandin E2, and caspase-3 activity in the cerebellar tissue.</p><p><strong>Conclusion: </strong>In conclusion, daflon confers neuroprotection against cisplatin-induced cerebellar neurotoxicity through the suppression of TLR4/NF-kB-mediated oxidative-inflammatory and apoptotic injury.</p>","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145572739","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Multifaceted anticancer activity of nafamostat mesylate in human fibrosarcoma: first evidence of mitochondrial apoptosis and suppressed MMP-2/-9 mRNA expression. 甲磺酸纳莫他酯在人纤维肉瘤中的多方面抗癌活性:线粒体凋亡和抑制MMP-2/-9 mRNA表达的第一个证据
IF 2.7 3区 医学 Q2 PHARMACOLOGY & PHARMACY Pub Date : 2025-11-19 DOI: 10.1186/s40360-025-01038-3
Cafer Yildirim, Filiz Bakar-Ates

Background: Fibrosarcoma is an aggressive soft tissue malignancy with limited therapeutic options, highlighting the need for novel treatment strategies. Nafamostat mesylate, a clinically approved serine protease inhibitor, has demonstrated anticancer effects in various tumor types, yet its impact on fibrosarcoma remains unexplored. This study aimed to investigate the cytotoxic, antimigratory, pro-apoptotic, and anti-invasive effects of nafamostat mesylate in human HT1080 fibrosarcoma cells.

Methods: HT1080 cells were treated with varying concentrations of nafamostat mesylate. Cell viability was assessed by MTT assay, migration by wound healing assay, and cell cycle distribution by flow cytometry. Apoptosis induction was evaluated using Annexin V binding assay, multicaspase activity, and mitochondrial membrane potential analysis. Additionally, mRNA expression levels of matrix metalloproteinases (MMPs), MMP-2 and MMP-9, were quantified by qRT-PCR.

Results: Nafamostat mesylate reduced HT1080 cell viability in a dose- and time-dependent manner and induced G2/M cell cycle arrest, indicating disruption of mitotic progression. Migration assays demonstrated suppression of cell motility. Apoptosis was confirmed through increased Annexin V positivity, elevated caspase activity, and mitochondrial depolarization, supporting caspase-dependent, mitochondria-mediated cell death. Furthermore, nafamostat mesylate treatment significantly downregulated MMP-2 and MMP-9 mRNA expression, suggesting inhibition of key enzymes responsible for extracellular matrix (ECM) degradation, invasion and metastasis.

Conclusions: This study provides the first evidence that nafamostat mesylate exerts multifaceted anticancer effects in HT1080 fibrosarcoma cells, targeting proliferation, migration, apoptosis, and invasion. These findings support the potential repurposing of nafamostat mesylate as a therapeutic agent for fibrosarcoma and warrant further preclinical investigations to evaluate its translational applicability.

背景:纤维肉瘤是一种侵袭性的软组织恶性肿瘤,治疗选择有限,强调需要新的治疗策略。甲磺酸纳莫司他是一种临床批准的丝氨酸蛋白酶抑制剂,已证明对多种肿瘤有抗癌作用,但其对纤维肉瘤的影响仍未研究。本研究旨在探讨甲磺酸那莫他酯对人HT1080纤维肉瘤细胞的细胞毒、抗迁移、促凋亡和抗侵袭作用。方法:用不同浓度甲磺酸那莫他处理HT1080细胞。MTT法测定细胞活力,伤口愈合法测定迁移量,流式细胞术测定细胞周期分布。通过Annexin V结合试验、多aspase活性和线粒体膜电位分析来评估细胞凋亡诱导。采用qRT-PCR方法检测基质金属蛋白酶(MMPs)、MMP-2和MMP-9的mRNA表达水平。结果:甲氨伐他酯以剂量和时间依赖的方式降低HT1080细胞活力,诱导G2/M细胞周期阻滞,表明有丝分裂进程中断。迁移试验显示细胞运动受到抑制。凋亡通过Annexin V阳性增加、caspase活性升高和线粒体去极化证实,支持caspase依赖、线粒体介导的细胞死亡。此外,甲磺酸纳莫司他显著下调了MMP-2和MMP-9 mRNA的表达,表明抑制了细胞外基质(ECM)降解、侵袭和转移的关键酶。结论:本研究首次证明甲磺酸那莫他酯对HT1080纤维肉瘤细胞具有多方面的抗癌作用,可靶向增殖、迁移、凋亡和侵袭。这些发现支持甲磺酸那莫他酯作为纤维肉瘤治疗剂的潜在用途,并需要进一步的临床前研究来评估其转化适用性。
{"title":"Multifaceted anticancer activity of nafamostat mesylate in human fibrosarcoma: first evidence of mitochondrial apoptosis and suppressed MMP-2/-9 mRNA expression.","authors":"Cafer Yildirim, Filiz Bakar-Ates","doi":"10.1186/s40360-025-01038-3","DOIUrl":"10.1186/s40360-025-01038-3","url":null,"abstract":"<p><strong>Background: </strong>Fibrosarcoma is an aggressive soft tissue malignancy with limited therapeutic options, highlighting the need for novel treatment strategies. Nafamostat mesylate, a clinically approved serine protease inhibitor, has demonstrated anticancer effects in various tumor types, yet its impact on fibrosarcoma remains unexplored. This study aimed to investigate the cytotoxic, antimigratory, pro-apoptotic, and anti-invasive effects of nafamostat mesylate in human HT1080 fibrosarcoma cells.</p><p><strong>Methods: </strong>HT1080 cells were treated with varying concentrations of nafamostat mesylate. Cell viability was assessed by MTT assay, migration by wound healing assay, and cell cycle distribution by flow cytometry. Apoptosis induction was evaluated using Annexin V binding assay, multicaspase activity, and mitochondrial membrane potential analysis. Additionally, mRNA expression levels of matrix metalloproteinases (MMPs), MMP-2 and MMP-9, were quantified by qRT-PCR.</p><p><strong>Results: </strong>Nafamostat mesylate reduced HT1080 cell viability in a dose- and time-dependent manner and induced G2/M cell cycle arrest, indicating disruption of mitotic progression. Migration assays demonstrated suppression of cell motility. Apoptosis was confirmed through increased Annexin V positivity, elevated caspase activity, and mitochondrial depolarization, supporting caspase-dependent, mitochondria-mediated cell death. Furthermore, nafamostat mesylate treatment significantly downregulated MMP-2 and MMP-9 mRNA expression, suggesting inhibition of key enzymes responsible for extracellular matrix (ECM) degradation, invasion and metastasis.</p><p><strong>Conclusions: </strong>This study provides the first evidence that nafamostat mesylate exerts multifaceted anticancer effects in HT1080 fibrosarcoma cells, targeting proliferation, migration, apoptosis, and invasion. These findings support the potential repurposing of nafamostat mesylate as a therapeutic agent for fibrosarcoma and warrant further preclinical investigations to evaluate its translational applicability.</p>","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":"26 1","pages":"194"},"PeriodicalIF":2.7,"publicationDate":"2025-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12628963/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145547881","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
BMC Pharmacology & Toxicology
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