{"title":"Unveiling the Influence of the Graded Impact of Cigarette Smoking Intensity on the Biochemical and Morphological Alterations of Erythrocytes.","authors":"Payel Biswas, Jyotirmoy Sikdar, Sutithi Dey, Baishali Basak, Sanjay Kumar Paul, Rajen Haldar","doi":"10.1007/s12013-025-01958-1","DOIUrl":"https://doi.org/10.1007/s12013-025-01958-1","url":null,"abstract":"","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":""},"PeriodicalIF":2.5,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145652914","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Correction: NanoBiT-based Analysis of Canine SOD1 Protein Dynamics: Understanding the Role of CCS and Ebselen Derivatives as Potential Therapeutics for Canine Degenerative Myelopathy.","authors":"Sakura Hirose, Yui Kobatake, Norihiro Tada, Mahmoud Kandeel, Akichika Itoh, Kentaro Oh-Hashi","doi":"10.1007/s12013-025-01795-2","DOIUrl":"10.1007/s12013-025-01795-2","url":null,"abstract":"","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"5557-5563"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144232885","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Type 2 diabetes (T2D) involves disrupted metabolism and immune responses, but mechanisms remain unclear. This study examines CD73 and its potential regulation by miR-30d-5p via the AMPK pathway. In db/db mice and clinical T2D patients, we observed significantly elevated hepatic CD73 expression inversely correlated with miR-30d-5p levels. siRNA-mediated CD73 silencing disrupted glucose-lipid metabolic stability by enhancing AMPK phosphorylation, suggesting CD73's regulatory role in energy homeostasis. Bioinformatics analysis identified CD73 as a putative target of miR-30 family members, which was experimentally validated through dual-luciferase reporter assays demonstrating miR-30d-5p's direct binding to two conserved sites within CD73's 3'-UTR. Notably, miR-30d-5p overexpression mimicked CD73 knockdown effects, downregulating key gluconeogenic enzymes while upregulating lipid oxidation markers. The inverse expression patterns of CD73 and miR-30d-5p in both murine models and human peripheral blood samples underscore their antagonistic relationship in T2D progression. These findings position the miR-30d-5p/CD73 axis as a critical regulator of hepatic AMPK-mediated metabolic processes, offering novel therapeutic targets. Our work bridges the gap between immune-modulatory pathways and metabolic regulation, proposing CD73 inhibition as a dual-action strategy for improving insulin sensitivity and mitigating diabetic complications. This mechanistic insight advances precision medicine approaches for T2D management by integrating epigenetic regulation with enzymatic metabolic control.
{"title":"MiR-30d-5p Modulates Glucose and Lipid Metabolism by Targeting CD73 through the AMPK Pathway.","authors":"Mingjie Song, Juan Wang, Wenli Mu, Yanwei Zheng, Yanjun Cai, Jing Liu, Yu Xia","doi":"10.1007/s12013-025-01815-1","DOIUrl":"10.1007/s12013-025-01815-1","url":null,"abstract":"<p><p>Type 2 diabetes (T2D) involves disrupted metabolism and immune responses, but mechanisms remain unclear. This study examines CD73 and its potential regulation by miR-30d-5p via the AMPK pathway. In db/db mice and clinical T2D patients, we observed significantly elevated hepatic CD73 expression inversely correlated with miR-30d-5p levels. siRNA-mediated CD73 silencing disrupted glucose-lipid metabolic stability by enhancing AMPK phosphorylation, suggesting CD73's regulatory role in energy homeostasis. Bioinformatics analysis identified CD73 as a putative target of miR-30 family members, which was experimentally validated through dual-luciferase reporter assays demonstrating miR-30d-5p's direct binding to two conserved sites within CD73's 3'-UTR. Notably, miR-30d-5p overexpression mimicked CD73 knockdown effects, downregulating key gluconeogenic enzymes while upregulating lipid oxidation markers. The inverse expression patterns of CD73 and miR-30d-5p in both murine models and human peripheral blood samples underscore their antagonistic relationship in T2D progression. These findings position the miR-30d-5p/CD73 axis as a critical regulator of hepatic AMPK-mediated metabolic processes, offering novel therapeutic targets. Our work bridges the gap between immune-modulatory pathways and metabolic regulation, proposing CD73 inhibition as a dual-action strategy for improving insulin sensitivity and mitigating diabetic complications. This mechanistic insight advances precision medicine approaches for T2D management by integrating epigenetic regulation with enzymatic metabolic control.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"4947-4960"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144566909","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-07-22DOI: 10.1007/s12013-025-01836-w
Mic Arun Edwin, Ramaiyan Velmurugan, Saraswati Patel
The rise in antifungal resistance underscores the need to explore novel bioactive compounds. This study investigates phytochemicals from Acalypha indica as potential inhibitors of lanosterol 14-alpha demethylase (CYP51) in Saccharomyces cerevisiae. A total of sixteen phytocompounds were evaluated using molecular docking, MM/GBSA binding free energy estimation, pharmacokinetic and toxicity predictions, and 200 ns molecular dynamics (MD) simulations. Among them, stigmasterol, aurantiamide, and beta-sitosterol showed strong binding affinities, comparable to standard drugs fluconazole and itraconazole. ADME analysis revealed good drug-likeness and gastrointestinal absorption for aurantiamide and 2-methylanthraquinone. ProTox-III predictions indicated low mutagenic and carcinogenic risks for most compounds, although aurantiamide may have nephrotoxic and respiratory toxicity concerns. Top ligands aurantiamide and stigmasterol were further subjected to MD simulations, which demonstrated stable RMSD, low RMSF, and well-maintained secondary structure, indicating strong interaction persistence and structural integrity. Ligand behaviour metrics (rGyr, SASA, MolSA, PSA, intra-HB) supported their binding stability. While aurantiamide exhibited an unfavourable binding energy (+228.37 kcal/mol), stigmasterol displayed a significantly favourable ΔG_bind (-93.36 kcal/mol). These findings suggest that stigmasterol and related phytochemicals hold promise as natural antifungal agents. However, further in vitro and in vivo validation, along with structure-activity relationship (SAR) optimization, is essential for clinical advancement.
{"title":"In Silico Evaluation of Acalypha indica Phytochemicals as Potential Antifungal Agents Targeting Saccharomyces cerevisiae Lanosterol 14-Alpha Demethylase.","authors":"Mic Arun Edwin, Ramaiyan Velmurugan, Saraswati Patel","doi":"10.1007/s12013-025-01836-w","DOIUrl":"10.1007/s12013-025-01836-w","url":null,"abstract":"<p><p>The rise in antifungal resistance underscores the need to explore novel bioactive compounds. This study investigates phytochemicals from Acalypha indica as potential inhibitors of lanosterol 14-alpha demethylase (CYP51) in Saccharomyces cerevisiae. A total of sixteen phytocompounds were evaluated using molecular docking, MM/GBSA binding free energy estimation, pharmacokinetic and toxicity predictions, and 200 ns molecular dynamics (MD) simulations. Among them, stigmasterol, aurantiamide, and beta-sitosterol showed strong binding affinities, comparable to standard drugs fluconazole and itraconazole. ADME analysis revealed good drug-likeness and gastrointestinal absorption for aurantiamide and 2-methylanthraquinone. ProTox-III predictions indicated low mutagenic and carcinogenic risks for most compounds, although aurantiamide may have nephrotoxic and respiratory toxicity concerns. Top ligands aurantiamide and stigmasterol were further subjected to MD simulations, which demonstrated stable RMSD, low RMSF, and well-maintained secondary structure, indicating strong interaction persistence and structural integrity. Ligand behaviour metrics (rGyr, SASA, MolSA, PSA, intra-HB) supported their binding stability. While aurantiamide exhibited an unfavourable binding energy (+228.37 kcal/mol), stigmasterol displayed a significantly favourable ΔG_bind (-93.36 kcal/mol). These findings suggest that stigmasterol and related phytochemicals hold promise as natural antifungal agents. However, further in vitro and in vivo validation, along with structure-activity relationship (SAR) optimization, is essential for clinical advancement.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"5197-5215"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144688493","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-07-21DOI: 10.1007/s12013-025-01841-z
Parisa Sadighara, Jamileh Salar-Amoli, Abbas Barin, Sher Ali, Carlos Augusto Fernandes Oliveira
The challenge in addressing methylmercury (MeHg) poisoning primarily lies in devising effective therapeutic strategies. In this study, we explore the potential cytoprotective effects of thiamine pyrophosphate (TPP) as a novel agent to alleviate MeHg-induced complications. Fetal fibroblast cells were exposed to 100 µM MeHg with varying concentrations of TPP (12.5-100 mM). Treated and control cells were analyzed for determination of DNA and protein contents, whereas glutathione and lipid peroxidation levels were measured as oxidative stress markers. TPP reduced the cellular lipid peroxidation and restored the intracellular glutathione levels altered by MeHg, also increasing the cell DNA content in the 12.5 mM TPP treatment group. TPP treatment led to enhanced cell survival, underscoring TPP's capacity to alleviate MeHg toxicity by improving the antioxidant status. Further studies on additional oxidative stress markers and apoptotic pathways are necessary to fully elucidate the scope and mechanisms of TPP's cytoprotective effects against MeHg toxicity. While the data in this trial highlight the potential of TPP as a novel therapeutic agent for individuals exposed to MeHg, clinical studies are required to confirm its protective efficacy aiming at developing future mitigation strategies.
解决甲基汞(MeHg)中毒的挑战主要在于制定有效的治疗策略。在这项研究中,我们探讨了焦磷酸硫胺素(TPP)作为一种新型药物的潜在细胞保护作用,以减轻甲基汞诱导的并发症。将胎儿成纤维细胞暴露于100µM MeHg和不同浓度的TPP (12.5-100 mM)中。分析处理细胞和对照细胞的DNA和蛋白质含量,并测量谷胱甘肽和脂质过氧化水平作为氧化应激标志物。在12.5 mM TPP处理组,TPP降低了细胞脂质过氧化,恢复了MeHg改变的细胞内谷胱甘肽水平,同时增加了细胞DNA含量。TPP处理导致细胞存活率提高,强调TPP通过改善抗氧化状态来减轻MeHg毒性的能力。需要进一步研究其他氧化应激标志物和凋亡途径,以充分阐明TPP对甲基汞毒性的细胞保护作用的范围和机制。虽然本试验中的数据强调了TPP作为MeHg暴露个体的新型治疗剂的潜力,但仍需要临床研究来确认其保护功效,以制定未来的缓解策略。
{"title":"Thiamine Mitigates the Toxicity of Methylmercury in Cultured Fetal Fibroblast Cell Lines.","authors":"Parisa Sadighara, Jamileh Salar-Amoli, Abbas Barin, Sher Ali, Carlos Augusto Fernandes Oliveira","doi":"10.1007/s12013-025-01841-z","DOIUrl":"10.1007/s12013-025-01841-z","url":null,"abstract":"<p><p>The challenge in addressing methylmercury (MeHg) poisoning primarily lies in devising effective therapeutic strategies. In this study, we explore the potential cytoprotective effects of thiamine pyrophosphate (TPP) as a novel agent to alleviate MeHg-induced complications. Fetal fibroblast cells were exposed to 100 µM MeHg with varying concentrations of TPP (12.5-100 mM). Treated and control cells were analyzed for determination of DNA and protein contents, whereas glutathione and lipid peroxidation levels were measured as oxidative stress markers. TPP reduced the cellular lipid peroxidation and restored the intracellular glutathione levels altered by MeHg, also increasing the cell DNA content in the 12.5 mM TPP treatment group. TPP treatment led to enhanced cell survival, underscoring TPP's capacity to alleviate MeHg toxicity by improving the antioxidant status. Further studies on additional oxidative stress markers and apoptotic pathways are necessary to fully elucidate the scope and mechanisms of TPP's cytoprotective effects against MeHg toxicity. While the data in this trial highlight the potential of TPP as a novel therapeutic agent for individuals exposed to MeHg, clinical studies are required to confirm its protective efficacy aiming at developing future mitigation strategies.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"5247-5253"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144673667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-07-31DOI: 10.1007/s12013-025-01852-w
Erkan Özbay, Ülkü Çömelekoğlu, Fatma Söğüt, Metin Yıldırım, Serkan Küççüktürk, Tuğce Duran, Mehmet Ali Karaselek, Hasan Hüseyin Doğan, Ümmühan Ünlü
{"title":"TRP Channel Inhibition and NF-κB Pathway Suppression in Human Ependymal Tumor cell-line by Achillea Biebersteinii Aqueous Extract.","authors":"Erkan Özbay, Ülkü Çömelekoğlu, Fatma Söğüt, Metin Yıldırım, Serkan Küççüktürk, Tuğce Duran, Mehmet Ali Karaselek, Hasan Hüseyin Doğan, Ümmühan Ünlü","doi":"10.1007/s12013-025-01852-w","DOIUrl":"10.1007/s12013-025-01852-w","url":null,"abstract":"","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"5321-5337"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144751993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
In cardiovascular research, melatonin has shown promise in exhibiting antifibrotic properties and modulating endoplasmic reticulum (ER) stress. However, the exact mechanism by which it influences myocardial fibrosis has not been fully clarified. Therefore, this research aimed to investigate the inhibitory effect of melatonin on the progression of myocardial fibrosis through a mechanism involving the BIP/PERK/CHOP signaling pathway, both in silico and in vivo experimental models. In in silico studies, molecular docking and molecular dynamics simulations were employed to predict the binding affinity of melatonin to ER stress arm proteins, BIP, and PERK. Following, in vivo experiments were carried out to confirm in silico analyses. In animal studies, rats were administered melatonin intraperitoneal (10 mg/kg per day) for 3 weeks, and on the 6th and 7th days, they were given isoproterenol at a dose of 170 mg/kg subcutaneous to estabilish myocardial fibrosis model. The morphological changes in cardiac tissue were assessed using hematoxylin and eosin (H&E) and Masson's trichrome staining. Additionally, the expression of BIP and CHOP, a key downstream target of the PERK pathway, was analyzed through real-time PCR and immunohistochemistry. In silico studies suggest melatonin interacts with BIP and PERK, demonstrating strong binding energy and forming a stable complex with both proteins. However, its affinity and stability with PERK are greater than with BIP. Furthermore, immunohistochemistry and qRT-PCR findings indicated that melatonin notably downregulated the expression of BIP and CHOP in the isoproterenol-induced cardiac fibrosis model. The strong binding affinity of melatonin for BIP and PERK, coupled with its impact on the downregulation of BIP and CHOP proteins in the isoproterenol-induced cardiac fibrosis model, suggests that melatonin's antifibrotic effects on myocardial tissue may be related to its ER stress inhibitory effects.
{"title":"Melatonin Ameliorates Isoproterenol-Induced Cardiac Fibrosis by Suppressing BIP/PERK/CHOP Signaling Pathways; Insights from In Silico and In vivo Studies.","authors":"Farkhondeh Pooresmaeil, Reza Jafari-Shakib, Behnam Hasannejad-Asl, Farhad Mashayekhi, Mojtaba Hedayati Ch, SeyyedMohammadTaghi Razavi-Toosi","doi":"10.1007/s12013-025-01847-7","DOIUrl":"10.1007/s12013-025-01847-7","url":null,"abstract":"<p><p>In cardiovascular research, melatonin has shown promise in exhibiting antifibrotic properties and modulating endoplasmic reticulum (ER) stress. However, the exact mechanism by which it influences myocardial fibrosis has not been fully clarified. Therefore, this research aimed to investigate the inhibitory effect of melatonin on the progression of myocardial fibrosis through a mechanism involving the BIP/PERK/CHOP signaling pathway, both in silico and in vivo experimental models. In in silico studies, molecular docking and molecular dynamics simulations were employed to predict the binding affinity of melatonin to ER stress arm proteins, BIP, and PERK. Following, in vivo experiments were carried out to confirm in silico analyses. In animal studies, rats were administered melatonin intraperitoneal (10 mg/kg per day) for 3 weeks, and on the 6th and 7th days, they were given isoproterenol at a dose of 170 mg/kg subcutaneous to estabilish myocardial fibrosis model. The morphological changes in cardiac tissue were assessed using hematoxylin and eosin (H&E) and Masson's trichrome staining. Additionally, the expression of BIP and CHOP, a key downstream target of the PERK pathway, was analyzed through real-time PCR and immunohistochemistry. In silico studies suggest melatonin interacts with BIP and PERK, demonstrating strong binding energy and forming a stable complex with both proteins. However, its affinity and stability with PERK are greater than with BIP. Furthermore, immunohistochemistry and qRT-PCR findings indicated that melatonin notably downregulated the expression of BIP and CHOP in the isoproterenol-induced cardiac fibrosis model. The strong binding affinity of melatonin for BIP and PERK, coupled with its impact on the downregulation of BIP and CHOP proteins in the isoproterenol-induced cardiac fibrosis model, suggests that melatonin's antifibrotic effects on myocardial tissue may be related to its ER stress inhibitory effects.</p>","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"5281-5295"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145022603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-08-08DOI: 10.1007/s12013-025-01855-7
Asal Shahmohammadi, Abdolreza Sabokrouh, Farrokh Modarresi
{"title":"Enhanced Anti-Leukemic Activity of Platinum Lamivudine Compared To Lamivudine Through Differential Gene Regulation and DNA Groove Binding.","authors":"Asal Shahmohammadi, Abdolreza Sabokrouh, Farrokh Modarresi","doi":"10.1007/s12013-025-01855-7","DOIUrl":"10.1007/s12013-025-01855-7","url":null,"abstract":"","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"5369-5386"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144797795","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-08-08DOI: 10.1007/s12013-025-01865-5
Akshad Balde, Rasool Abdul Nazeer
{"title":"Synergistic Effects of LPS and MSU on NF-κB/NLRP3-mediated Inflammation in Fibroblast Cells.","authors":"Akshad Balde, Rasool Abdul Nazeer","doi":"10.1007/s12013-025-01865-5","DOIUrl":"10.1007/s12013-025-01865-5","url":null,"abstract":"","PeriodicalId":510,"journal":{"name":"Cell Biochemistry and Biophysics","volume":" ","pages":"5499-5512"},"PeriodicalIF":2.5,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144797796","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}