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.
{"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}
{"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}
{"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}
{"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}
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}
Pub Date : 2025-11-21DOI: 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.
{"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}
Pub Date : 2025-11-21DOI: 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.
{"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}
Pub Date : 2025-11-19DOI: 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.
{"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}