Pub Date : 2025-12-11DOI: 10.1016/j.ejphar.2025.178472
Xuanrui Xiong , Qiang Li , Yuanyuan Zhang , Pan Liu , Yuxu Pan , Qiongtao Song
Copper is a vital trace element for all living organisms and plays an important role in numerous physiological functions, including the formation of mitochondrial respiratory chain complexes, antioxidant defense, and signal transduction. However, excess copper can cause cellular toxicity and initiate a form of cell death that is characterized by the aggregation of lipoylated proteins and a reduction in Fe-S cluster proteins. This series of events can culminate in mitochondrial process of respiratory dysfunction known as cuproptosis. Excessive copper can also inhibit the ubiquitin–proteasome system, which results in the accumulation of harmful proteins and a vicious cycle of Fenton and Haber–Weiss reactions that trigger oxidative stress and cellular damage. The eye, particularly the retina, is one of the most energy-dependent tissues in the body and has an extraordinary dependence on mitochondrial function. Dysregulated copper-ion levels can lead to mitochondrial dysfunction, which can cause various ocular diseases, including uveal melanoma, age-related macular degeneration, and diabetic retinopathy. Therefore, the relationship between copper and ocular diseases provides promising research opportunities. This review summarizes recent research findings on copper metabolism, cuproptosis, and their implications in ocular diseases. It also introduces potential therapeutic approaches for related diseases, including copper chelation therapy, copper ionophores and nanomedicine, and genetic treatment strategies.
{"title":"Exploring copper metabolism and cuproptosis, and their implications in ocular diseases","authors":"Xuanrui Xiong , Qiang Li , Yuanyuan Zhang , Pan Liu , Yuxu Pan , Qiongtao Song","doi":"10.1016/j.ejphar.2025.178472","DOIUrl":"10.1016/j.ejphar.2025.178472","url":null,"abstract":"<div><div>Copper is a vital trace element for all living organisms and plays an important role in numerous physiological functions, including the formation of mitochondrial respiratory chain complexes, antioxidant defense, and signal transduction. However, excess copper can cause cellular toxicity and initiate a form of cell death that is characterized by the aggregation of lipoylated proteins and a reduction in Fe-S cluster proteins. This series of events can culminate in mitochondrial process of respiratory dysfunction known as cuproptosis. Excessive copper can also inhibit the ubiquitin–proteasome system, which results in the accumulation of harmful proteins and a vicious cycle of Fenton and Haber–Weiss reactions that trigger oxidative stress and cellular damage. The eye, particularly the retina, is one of the most energy-dependent tissues in the body and has an extraordinary dependence on mitochondrial function. Dysregulated copper-ion levels can lead to mitochondrial dysfunction, which can cause various ocular diseases, including uveal melanoma, age-related macular degeneration, and diabetic retinopathy. Therefore, the relationship between copper and ocular diseases provides promising research opportunities. This review summarizes recent research findings on copper metabolism, cuproptosis, and their implications in ocular diseases. It also introduces potential therapeutic approaches for related diseases, including copper chelation therapy, copper ionophores and nanomedicine, and genetic treatment strategies.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178472"},"PeriodicalIF":4.7,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145735089","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 : 2025-12-10DOI: 10.1016/j.ejphar.2025.178438
Xiaoyao Chang , Xiaoyang Wang , Mengyun Wang , Yuanhao Tan , Yawen Xie , Dangran Li , Hailin Ma , Fangfang Cai , Hongqin Zhuang , Zi-Chun Hua
Metformin, a widely used agent for type 2 diabetes treatment, has been found to reduce the risk of cancer in patients with diabetes in recent years. In preclinical studies, researchers found that metformin, when used alone, exhibits anti-tumor effects both in vitro and in vivo. However, the anti-tumor mechanisms of metformin remain to be further explored. In this study, we found that metformin alkalized endo-lysosome pH and inhibited autophagy by targeting the endosomal transmembrane protein NHE6 (Na+-H+ Exchanger 6), thereby inhibiting melanoma cell growth and survival. In contrast, the knockout of NHE6 in B16F10 cells acidified the endo-lysosomal pH and activated autophagy, which in turn reversed the metformin-induced pH and autophagy regulation and blunted the anti-tumor effects of metformin both in vitro and in vivo. Interestingly, knockout of NHE6 also suppressed B16F10 cell growth and survival. However, NHE6 could be rescued by restoration of endo-lysosome pH, suggesting that activation or inhibition of autophagy by pH interference might be detrimental to the fate of B16F10 cells. Taken together, we conclude that metformin could suppress melanoma progression by inhibiting autophagy based on endo-lysosome pH regulation. In addition, we also found the potential role of NHE6 in the progression of melanoma, which is worth further study as novel theoretical evidence for future melanoma pathogenesis and targeted therapy research.
{"title":"Metformin's hidden power: Unveiling its role in melanoma suppression through endo-lysosome pH and autophagy regulation","authors":"Xiaoyao Chang , Xiaoyang Wang , Mengyun Wang , Yuanhao Tan , Yawen Xie , Dangran Li , Hailin Ma , Fangfang Cai , Hongqin Zhuang , Zi-Chun Hua","doi":"10.1016/j.ejphar.2025.178438","DOIUrl":"10.1016/j.ejphar.2025.178438","url":null,"abstract":"<div><div>Metformin, a widely used agent for type 2 diabetes treatment, has been found to reduce the risk of cancer in patients with diabetes in recent years. In preclinical studies, researchers found that metformin, when used alone, exhibits anti-tumor effects both <em>in vitro</em> and <em>in vivo</em>. However, the anti-tumor mechanisms of metformin remain to be further explored. In this study, we found that metformin alkalized endo-lysosome pH and inhibited autophagy by targeting the endosomal transmembrane protein NHE6 (Na<sup>+</sup>-H<sup>+</sup> Exchanger 6), thereby inhibiting melanoma cell growth and survival. In contrast, the knockout of NHE6 in B16F10 cells acidified the endo-lysosomal pH and activated autophagy, which in turn reversed the metformin-induced pH and autophagy regulation and blunted the anti-tumor effects of metformin both <em>in vitro</em> and <em>in vivo</em>. Interestingly, knockout of NHE6 also suppressed B16F10 cell growth and survival. However, NHE6 could be rescued by restoration of endo-lysosome pH, suggesting that activation or inhibition of autophagy by pH interference might be detrimental to the fate of B16F10 cells. Taken together, we conclude that metformin could suppress melanoma progression by inhibiting autophagy based on endo-lysosome pH regulation. In addition, we also found the potential role of NHE6 in the progression of melanoma, which is worth further study as novel theoretical evidence for future melanoma pathogenesis and targeted therapy research.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178438"},"PeriodicalIF":4.7,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145735042","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 : 2025-12-10DOI: 10.1016/j.ejphar.2025.178459
Tashinga Walter Matiki , Mohd Zahoor Ul Haq Shah , Lin Yin , Rui Liu , Kejing Zhu , Zhongliang Lin , Jianzhong Sheng , Huang Hefeng
Obesity-induced reproductive dysfunction is driven by chronic low-grade inflammation and metabolic dysregulation. The potential modulatory effect of GLP-1RAs on the interplay mechanisms of inflammation and metabolic dysregulation in obesity-induced reproductive dysfunction remains unclear. Hence, we investigated the effect of semaglutide on reproductive function via the SIRT-associated pathway in obese female mice.
Female mice were divided into three groups: control-normal diet (NC), obesity-high fat diet (HFD), and intervention-high fat diet + semaglutide (HS). Mice body weight, composition and metabolic response to glucose, insulin, and pyruvate were evaluated. Systemic and ovarian inflammatory cytokines (IL-1β, NF-κB, IL-6, TNF-α), reproductive hormones (P4, E2, FSH, LH), and ovarian expression of SIRT1, SIRT6, FOXO1, FOXO3a, NRF1, IRS1, iNOS, eNOS, p27 KIP1, and PTEN were evaluated. The ovarian tissues were analyzed for structural changes, lipid accumulation, oxidative stress, and follicular development. Oocyte's mitochondrial function, DNA damage, lipid and ROS levels, and fertility tests were assessed.
Adiposity and metabolic dysregulation, mainly insulin resistance and IRS1 disruption, were observed in the obese group. The HFD group showed higher NF-κB-associated pro-inflammatory cytokine expressions, oxidative stress, FOXO1 and iNOS expression, reduced SIRT1, SIRT6, FOXO3a, NRF1, eNOS levels, and dysregulated p27 KIP1/PTEN colocalization. These observed changes were accompanied by reduced P4 and E2, elevated FSH and LH, disrupted ovarian morphology, ovarian and oocyte lipid accumulation, oocyte DNA damage, and reduced fertility outcomes in the HFD group. Semaglutide treatment alleviated the dysfunctions observed in the obese group.
Our findings demonstrated that GLP-1RAs (semaglutide) have potential therapeutic effects on obesity-induced reproductive dysfunctions by modulating the interplay mechanisms of inflammation and metabolic dysregulation via the SIRT-associated pathway.
{"title":"GLP-1RA partially alleviates obesity-induced reproductive dysfunction driven by the interplay mechanisms of inflammation and metabolic dysregulation via the SIRT-associated pathway","authors":"Tashinga Walter Matiki , Mohd Zahoor Ul Haq Shah , Lin Yin , Rui Liu , Kejing Zhu , Zhongliang Lin , Jianzhong Sheng , Huang Hefeng","doi":"10.1016/j.ejphar.2025.178459","DOIUrl":"10.1016/j.ejphar.2025.178459","url":null,"abstract":"<div><div>Obesity-induced reproductive dysfunction is driven by chronic low-grade inflammation and metabolic dysregulation. The potential modulatory effect of GLP-1RAs on the interplay mechanisms of inflammation and metabolic dysregulation in obesity-induced reproductive dysfunction remains unclear. Hence, we investigated the effect of semaglutide on reproductive function via the SIRT-associated pathway in obese female mice.</div><div>Female mice were divided into three groups: control-normal diet (NC), obesity-high fat diet (HFD), and intervention-high fat diet + semaglutide (HS). Mice body weight, composition and metabolic response to glucose, insulin, and pyruvate were evaluated. Systemic and ovarian inflammatory cytokines (IL-1β, NF-κB, IL-6, TNF-α), reproductive hormones (P4, E2, FSH, LH), and ovarian expression of SIRT1, SIRT6, FOXO1, FOXO3a, NRF1, IRS1, iNOS, eNOS, p27 KIP1, and PTEN were evaluated. The ovarian tissues were analyzed for structural changes, lipid accumulation, oxidative stress, and follicular development. Oocyte's mitochondrial function, DNA damage, lipid and ROS levels, and fertility tests were assessed.</div><div>Adiposity and metabolic dysregulation, mainly insulin resistance and IRS1 disruption, were observed in the obese group. The HFD group showed higher NF-κB-associated pro-inflammatory cytokine expressions, oxidative stress, FOXO1 and iNOS expression, reduced SIRT1, SIRT6, FOXO3a, NRF1, eNOS levels, and dysregulated p27 KIP1/PTEN colocalization. These observed changes were accompanied by reduced P4 and E2, elevated FSH and LH, disrupted ovarian morphology, ovarian and oocyte lipid accumulation, oocyte DNA damage, and reduced fertility outcomes in the HFD group. Semaglutide treatment alleviated the dysfunctions observed in the obese group.</div><div>Our findings demonstrated that GLP-1RAs (semaglutide) have potential therapeutic effects on obesity-induced reproductive dysfunctions by modulating the interplay mechanisms of inflammation and metabolic dysregulation via the SIRT-associated pathway.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178459"},"PeriodicalIF":4.7,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145735041","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 : 2025-12-10DOI: 10.1016/j.ejphar.2025.178457
Tian Zhao , Quanxin Liu , Jianzhou Chen , Wenjing Xie , Qinghua Jin , Bin Xiao
The integrated stress response (ISR) has been implicated in cognitive decline associated with ageing and neurodegenerative diseases. Pharmacological inhibition of the ISR using the small-molecule ISRIB has demonstrated promising neuroprotective effects in several preclinical models. However, its potential therapeutic value in vascular cognitive impairment (VCI) remains largely unexplored. Here, we established a modified permanent bilateral carotid occlusion (2-VO) rat model of VCI and investigated the therapeutic potential of the ISRIB via microinjection in hippocampal dentate gyrus (DG). VCI rats exhibited elevated expression of vascular endothelial growth factor (VEGF), cluster of differentiation 34 (CD34), ionized calcium-binding adapter molecule 1 (Iba1), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6), indicating successful establishment of the model. Behavioral assessments revealed that VCI rats exhibited impaired spatial, working, and recognition memory. Bioinformatic analysis highlighted ISR pathway activation in VCI. Furthermore, elevated phosphorylated eukaryotic initiation factor 2 alpha (p-eIF2α) and activating transcription factor 4 (ATF4) protein levels in the DG confirmed ISR activation in the DG of VCI rats. VCI also reduced neuronal integrity, as evidenced by decreased Nissl body density. ISRIB treatment significantly improved cognitive performance, suppressed ATF4 expression, enhanced puromycin-labeled protein synthesis, and restored phosphorylated cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) signaling. Notably, ISRIB increased c-fos activation and upregulated synaptophysin and postsynaptic density protein 95 (PSD95) expression in the DG of VCI rats, indicating enhanced neuronal activity and synaptic function. Our results indicate that ISR activation contributes to hippocampal-dependent memory impairment in VCI. ISRIB effectively restores synaptic function and cognition, underscoring its therapeutic value and translational potential in treating VCI.
{"title":"Targeting the integrated stress response with ISRIB enhances CREB/BDNF signaling and attenuates cognitive deficits in a rat model of vascular cognitive impairment","authors":"Tian Zhao , Quanxin Liu , Jianzhou Chen , Wenjing Xie , Qinghua Jin , Bin Xiao","doi":"10.1016/j.ejphar.2025.178457","DOIUrl":"10.1016/j.ejphar.2025.178457","url":null,"abstract":"<div><div>The integrated stress response (ISR) has been implicated in cognitive decline associated with ageing and neurodegenerative diseases. Pharmacological inhibition of the ISR using the small-molecule ISRIB has demonstrated promising neuroprotective effects in several preclinical models. However, its potential therapeutic value in vascular cognitive impairment (VCI) remains largely unexplored. Here, we established a modified permanent bilateral carotid occlusion (2-VO) rat model of VCI and investigated the therapeutic potential of the ISRIB <em>via</em> microinjection in hippocampal dentate gyrus (DG). VCI rats exhibited elevated expression of vascular endothelial growth factor (VEGF), cluster of differentiation 34 (CD34), ionized calcium-binding adapter molecule 1 (Iba1), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6), indicating successful establishment of the model. Behavioral assessments revealed that VCI rats exhibited impaired spatial, working, and recognition memory. Bioinformatic analysis highlighted ISR pathway activation in VCI. Furthermore, elevated phosphorylated eukaryotic initiation factor 2 alpha (p-eIF2α) and activating transcription factor 4 (ATF4) protein levels in the DG confirmed ISR activation in the DG of VCI rats. VCI also reduced neuronal integrity, as evidenced by decreased Nissl body density. ISRIB treatment significantly improved cognitive performance, suppressed ATF4 expression, enhanced puromycin-labeled protein synthesis, and restored phosphorylated cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) signaling. Notably, ISRIB increased c-fos activation and upregulated synaptophysin and postsynaptic density protein 95 (PSD95) expression in the DG of VCI rats, indicating enhanced neuronal activity and synaptic function. Our results indicate that ISR activation contributes to hippocampal-dependent memory impairment in VCI. ISRIB effectively restores synaptic function and cognition, underscoring its therapeutic value and translational potential in treating VCI.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178457"},"PeriodicalIF":4.7,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145735092","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 : 2025-12-10DOI: 10.1016/j.ejphar.2025.178468
Yue Li , Shoumeng Han , Tingting Xie , Kun Song , Weiye Zeng , Hanbing Wang , Chengyuan Hu , Zhijing Zhang , Kenji Hashimoto , Li Ma , Haihui Xie
Depression is a major global health burden, and current treatments are limited by delayed onset and incomplete efficacy, highlighting the need for novel, mechanism-based therapies. Chronic restraint stress (CRS) induces behavioral, hormonal, and synaptic changes relevant to depression, but the role of adiponectin signaling remains unclear. Here, we examined whether the adiponectin receptor agonist AdipoRon exerts antidepressant-like effects via brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signaling in mice subjected to 14 days of CRS. CRS produced anxiety- and depression-like behaviors, elevated plasma corticosterone, reduced circulating adiponectin, and selectively decreased hippocampal adiponectin and adiponectin receptor 2 (AdipoR2), accompanied by reduced PSD-95 and GluA1 in CA3 and the dentate gyrus (DG). AdipoRon treatment (20 mg/kg, days 8–14) prevented behavioral deficits, normalized corticosterone and adiponectin levels, and restored hippocampal AdipoR2, PSD-95, and GluA1 expression in CA3 and DG. AdipoRon also reversed CRS-induced decreases in hippocampal phosphorylated AMPK (p-AMPK), PPARα, BDNF, and phosphorylated TrkB (p-TrkB), with p-AMPK/AMPK and PPARα levels positively correlating with BDNF. Immunofluorescence confirmed BDNF recovery in CA3 and DG. Importantly, pretreatment with the TrkB antagonist ANA-12 abolished the behavioral, hormonal, and molecular effects of AdipoRon, indicating that its actions require BDNF–TrkB activation. These findings suggest that AdipoRon mitigates CRS-induced deficits via hippocampal AdipoR2–AMPK–PPARα–BDNF–TrkB signaling and highlight AdipoR2 as a promising target for depression therapy under chronic stress.
{"title":"AdipoRon ameliorates anxiety- and depression-like behaviors in chronic restraint-stressed mice via AMPK-PPARα-BDNF-TrkB signaling","authors":"Yue Li , Shoumeng Han , Tingting Xie , Kun Song , Weiye Zeng , Hanbing Wang , Chengyuan Hu , Zhijing Zhang , Kenji Hashimoto , Li Ma , Haihui Xie","doi":"10.1016/j.ejphar.2025.178468","DOIUrl":"10.1016/j.ejphar.2025.178468","url":null,"abstract":"<div><div>Depression is a major global health burden, and current treatments are limited by delayed onset and incomplete efficacy, highlighting the need for novel, mechanism-based therapies. Chronic restraint stress (CRS) induces behavioral, hormonal, and synaptic changes relevant to depression, but the role of adiponectin signaling remains unclear. Here, we examined whether the adiponectin receptor agonist AdipoRon exerts antidepressant-like effects via brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signaling in mice subjected to 14 days of CRS. CRS produced anxiety- and depression-like behaviors, elevated plasma corticosterone, reduced circulating adiponectin, and selectively decreased hippocampal adiponectin and adiponectin receptor 2 (AdipoR2), accompanied by reduced PSD-95 and GluA1 in CA3 and the dentate gyrus (DG). AdipoRon treatment (20 mg/kg, days 8–14) prevented behavioral deficits, normalized corticosterone and adiponectin levels, and restored hippocampal AdipoR2, PSD-95, and GluA1 expression in CA3 and DG. AdipoRon also reversed CRS-induced decreases in hippocampal phosphorylated AMPK (p-AMPK), PPARα, BDNF, and phosphorylated TrkB (p-TrkB), with p-AMPK/AMPK and PPARα levels positively correlating with BDNF. Immunofluorescence confirmed BDNF recovery in CA3 and DG. Importantly, pretreatment with the TrkB antagonist ANA-12 abolished the behavioral, hormonal, and molecular effects of AdipoRon, indicating that its actions require BDNF–TrkB activation. These findings suggest that AdipoRon mitigates CRS-induced deficits via hippocampal AdipoR2–AMPK–PPARα–BDNF–TrkB signaling and highlight AdipoR2 as a promising target for depression therapy under chronic stress.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178468"},"PeriodicalIF":4.7,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145735044","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 : 2025-12-09DOI: 10.1016/j.ejphar.2025.178462
Haichuan Miao , Baolong Liu , Xinfang Pan , Zhihua Cheng , Qizhen Xue , Dongda Han , Zhongshen Yu , Liwei Zhang , Xizhuang Bai
Objective
Osteosarcoma (OS), the most common primary malignant bone tumor, exhibits poor prognosis, underscoring the need for new therapies. Juglone, a natural naphthoquinone, shows therapeutic promise; however, the effects of juglone on OS and underlying mechanisms remain to be elucidated.
Materials and methods
In vitro experiments were validated using U2OS and HOS cell lines. Network pharmacology predicted juglone-OS targets, followed by protein-protein interaction (PPI) network construction and GO/KEGG enrichment analysis. Transcriptomics analysis was further confirmed. Molecular docking was performed using AutoDock Tools and Discovery Studio. Molecular dynamics simulations (MD) using GROMACS confirmed the stability of the juglone-target complex. In vivo experiments clarified the effect of juglone in vivo. ADME/T was profiled using Swiss and Protox.
Results
Juglone inhibited OS cell viability, promoted apoptosis, suppressed proliferation and migration, and induced G2/M arrest. A total of 234 targets were identified; PPI highlighted key interactors (TP53, AKT1, BCL2, CTNNB1, STAT3). Enrichment analyses revealed associations with oxidative stress and PI3K/AKT pathways. Molecular docking showed high-affinity binding to PI3K and AKT1, confirmed stable by molecular dynamics. Mechanistically, juglone inhibits OS via the ROS/PI3K/AKT pathway. In vivo, it potently inhibited tumor growth in xenograft models with short-term biosafety. ADME/T profiling indicated favorable pharmacokinetics but potential toxicity risks.
Conclusion
This study first demonstrates juglone's anti-OS efficacy via the ROS/PI3K/AKT pathway. In vivo studies confirm potent tumor suppression with short-term safety, supporting its clinical promise for OS treatment.
{"title":"Integrated strategies of network pharmacology, transcriptomics, and computational and experimental validation reveal the anti-osteosarcoma effects of juglone via the ROS/PI3K/AKT pathway","authors":"Haichuan Miao , Baolong Liu , Xinfang Pan , Zhihua Cheng , Qizhen Xue , Dongda Han , Zhongshen Yu , Liwei Zhang , Xizhuang Bai","doi":"10.1016/j.ejphar.2025.178462","DOIUrl":"10.1016/j.ejphar.2025.178462","url":null,"abstract":"<div><h3>Objective</h3><div>Osteosarcoma (OS), the most common primary malignant bone tumor, exhibits poor prognosis, underscoring the need for new therapies. Juglone, a natural naphthoquinone, shows therapeutic promise; however, the effects of juglone on OS and underlying mechanisms remain to be elucidated.</div></div><div><h3>Materials and methods</h3><div>In vitro experiments were validated using U2OS and HOS cell lines. Network pharmacology predicted juglone-OS targets, followed by protein-protein interaction (PPI) network construction and GO/KEGG enrichment analysis. Transcriptomics analysis was further confirmed. Molecular docking was performed using AutoDock Tools and Discovery Studio. Molecular dynamics simulations (MD) using GROMACS confirmed the stability of the juglone-target complex. In vivo experiments clarified the effect of juglone in vivo. ADME/T was profiled using Swiss and Protox.</div></div><div><h3>Results</h3><div>Juglone inhibited OS cell viability, promoted apoptosis, suppressed proliferation and migration, and induced G2/M arrest. A total of 234 targets were identified; PPI highlighted key interactors (TP53, AKT1, BCL2, CTNNB1, STAT3). Enrichment analyses revealed associations with oxidative stress and PI3K/AKT pathways. Molecular docking showed high-affinity binding to PI3K and AKT1, confirmed stable by molecular dynamics. Mechanistically, juglone inhibits OS via the ROS/PI3K/AKT pathway. In vivo, it potently inhibited tumor growth in xenograft models with short-term biosafety. ADME/T profiling indicated favorable pharmacokinetics but potential toxicity risks.</div></div><div><h3>Conclusion</h3><div>This study first demonstrates juglone's anti-OS efficacy via the ROS/PI3K/AKT pathway. In vivo studies confirm potent tumor suppression with short-term safety, supporting its clinical promise for OS treatment.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178462"},"PeriodicalIF":4.7,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145735093","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 : 2025-12-09DOI: 10.1016/j.ejphar.2025.178463
Song Xue , Yuange Li , Yin Zhang , Qiang Zhang , Sifei Yu , Lei Xie , Yu Liang
Spinal cord ischemia-reperfusion injury (SCIRI) leads to acute neurological deficits primarily driven by oxidative stress and apoptotic cell death. The potential of exogenous nicotinamide adenine dinucleotide (NAD+) to confer acute neuroprotection, as well as the underlying mechanisms, remains poorly understood. Network pharmacology analysis implicated the PI3K/AKT signaling pathway as a potential mediator of NAD+ effects. To investigate this, we assessed NAD+ in both in vivo (rodent SCIRI model; 75 mg/kg, intraperitoneal administration at reperfusion) and in vitro (oxygen-glucose deprivation/reperfusion, OGD/R; 32 μM) settings. Functional recovery was evaluated using the BBB locomotor rating scale, while histopathological changes, apoptosis (via TUNEL staining), oxidative stress markers (SOD, MDA, ROS), and key apoptotic proteins (Bcl-2/Bax, cleaved caspase-3) were analyzed alongside PI3K/AKT pathway activation. Pharmacological inhibition with LY294002 was employed to validate pathway involvement. Our results demonstrate that NAD+ significantly improved motor function recovery, attenuated histological damage, and reduced both apoptosis and oxidative stress following SCIRI. Consistent cytoprotection was observed in the OGD/R model with 32 μM NAD+. Mechanistically, NAD+ restored phosphorylation levels of PI3K and AKT and favorably modulated apoptotic regulators—effects partially reversed by LY294002, confirming the critical role of the PI3K/AKT axis. These findings provide mechanistic evidence that exogenous NAD+ exerts acute neuroprotection during the early reperfusion phase, mediated through PI3K/AKT pathway activation and subsequent suppression of oxidative stress and apoptosis.
{"title":"Nicotinamide adenine dinucleotide attenuates neuronal apoptosis in spinal cord ischemia-reperfusion injury via the activation of PI3K/AKT pathway","authors":"Song Xue , Yuange Li , Yin Zhang , Qiang Zhang , Sifei Yu , Lei Xie , Yu Liang","doi":"10.1016/j.ejphar.2025.178463","DOIUrl":"10.1016/j.ejphar.2025.178463","url":null,"abstract":"<div><div>Spinal cord ischemia-reperfusion injury (SCIRI) leads to acute neurological deficits primarily driven by oxidative stress and apoptotic cell death. The potential of exogenous nicotinamide adenine dinucleotide (NAD<sup>+</sup>) to confer acute neuroprotection, as well as the underlying mechanisms, remains poorly understood. Network pharmacology analysis implicated the PI3K/AKT signaling pathway as a potential mediator of NAD<sup>+</sup> effects. To investigate this, we assessed NAD<sup>+</sup> in both in vivo (rodent SCIRI model; 75 mg/kg, intraperitoneal administration at reperfusion) and in vitro (oxygen-glucose deprivation/reperfusion, OGD/R; 32 μM) settings. Functional recovery was evaluated using the BBB locomotor rating scale, while histopathological changes, apoptosis (via TUNEL staining), oxidative stress markers (SOD, MDA, ROS), and key apoptotic proteins (Bcl-2/Bax, cleaved caspase-3) were analyzed alongside PI3K/AKT pathway activation. Pharmacological inhibition with LY294002 was employed to validate pathway involvement. Our results demonstrate that NAD<sup>+</sup> significantly improved motor function recovery, attenuated histological damage, and reduced both apoptosis and oxidative stress following SCIRI. Consistent cytoprotection was observed in the OGD/R model with 32 μM NAD<sup>+</sup>. Mechanistically, NAD<sup>+</sup> restored phosphorylation levels of PI3K and AKT and favorably modulated apoptotic regulators—effects partially reversed by LY294002, confirming the critical role of the PI3K/AKT axis. These findings provide mechanistic evidence that exogenous NAD<sup>+</sup> exerts acute neuroprotection during the early reperfusion phase, mediated through PI3K/AKT pathway activation and subsequent suppression of oxidative stress and apoptosis.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178463"},"PeriodicalIF":4.7,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145735090","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 : 2025-12-08DOI: 10.1016/j.ejphar.2025.178389
Tao Liu , Yuhan Sun , Yangchun Liu , Xiantao Wang , Jian Xie , Jin Shu , Tao Li , Haoliang Li , Lang Li
Coronary microembolization (CME) is associated with an adverse prognosis in patients with acute coronary syndrome. However, the roles of long noncoding RNAs (lncRNAs) driven by super-enhancers (SEs) in the heart following CME have not been well elucidated. Here, we identified SE-associated lncRNA SHC (SE-regulated lncRNA highly expressed in CME) from histone H3 lysine-27 acetylation (H3K27ac) ChIP-seq data derived from rat hearts subjected to CME. A significant upregulation of lncRNA SHC was found in heart tissue following CME and Erastin-induced H9c2 cardiomyocytes. High levels of lncRNA SHC contributed to cardiac ferroptosis and subsequent myocardial injury following CME, while lncRNA SHC knockdown reversed this effect. Mechanistically, transcription factor specific protein 1 (SP1) binds to the SE at the lncRNA SHC locus and induces transcription. Furthermore, lncRNA SHC acted as a sponge for miR-26b-3p to promote oxidized low-density lipoprotein receptor 1 (Olr1) expression, which partially accounted for ferroptosis-dependent myocardial injury. In conclusion, these data suggest that the lncRNA SHC, driven by SE, contributes to cardiac ferroptosis and myocardial injury by targeting the miR-26b-3p/Olr1 cascade, and thus may be a promising therapeutic target to improve the prognosis of patients with CME.
{"title":"Inhibition of super-enhancer-driven lncRNA SHC alleviates coronary microembolization-induced myocardial injury by inhibiting cardiac ferroptosis","authors":"Tao Liu , Yuhan Sun , Yangchun Liu , Xiantao Wang , Jian Xie , Jin Shu , Tao Li , Haoliang Li , Lang Li","doi":"10.1016/j.ejphar.2025.178389","DOIUrl":"10.1016/j.ejphar.2025.178389","url":null,"abstract":"<div><div>Coronary microembolization (CME) is associated with an adverse prognosis in patients with acute coronary syndrome. However, the roles of long noncoding RNAs (lncRNAs) driven by super-enhancers (SEs) in the heart following CME have not been well elucidated. Here, we identified SE-associated lncRNA SHC (SE-regulated lncRNA highly expressed in CME) from histone H3 lysine-27 acetylation (H3K27ac) ChIP-seq data derived from rat hearts subjected to CME. A significant upregulation of lncRNA SHC was found in heart tissue following CME and Erastin-induced H9c2 cardiomyocytes. High levels of lncRNA SHC contributed to cardiac ferroptosis and subsequent myocardial injury following CME, while lncRNA SHC knockdown reversed this effect. Mechanistically, transcription factor specific protein 1 (SP1) binds to the SE at the lncRNA SHC locus and induces transcription. Furthermore, lncRNA SHC acted as a sponge for miR-26b-3p to promote oxidized low-density lipoprotein receptor 1 (Olr1) expression, which partially accounted for ferroptosis-dependent myocardial injury. In conclusion, these data suggest that the lncRNA SHC, driven by SE, contributes to cardiac ferroptosis and myocardial injury by targeting the miR-26b-3p/Olr1 cascade, and thus may be a promising therapeutic target to improve the prognosis of patients with CME.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178389"},"PeriodicalIF":4.7,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721502","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 : 2025-12-08DOI: 10.1016/j.ejphar.2025.178458
Zhongyu Ma , Jiawen Hao , Zhaohui Yang , Miao Zhang , Yuan Peng , Jizhao Xin , Yuanting Yang , Mengke Wu , Xuewei Yin , Dadong Guo , Hongsheng Bi
Objective
This study aimed to investigate the role of leukocyte immunoglobulin-like receptor B2 (Lilrb2) in suppressing scleral remodeling in a guinea pig model of lens-induced myopia (LIM), with a focus on its ability to modulate the intraocular inflammatory microenvironment.
Methods
An LIM model was established in guinea pigs using −6.0 D lenses. Animals received an intravitreal injection of an adeno-associated virus overexpressing Lilrb2 (AAV-Lilrb2). Ocular biometric parameters were monitored. The activity of the MEK-ERK-P65 signaling pathway in the retina and the protein levels of MMP-2, collagen I, and α-SMA in the sclera were assessed using qPCR and Western blot. Macrophage polarization states were evaluated via multiplex immunofluorescence and Western blot analysis of iNOS (M1 marker) and ARG1 (M2 marker). Structural changes in the sclera were examined by H&E staining, while collagen organization and fibrosis were evaluated using Masson staining and immunofluorescence.
Results
Compared with the normal control (NC) group, the LIM group exhibited significant axial elongation and a myopic shift in refractive error. In the retina, LIM promoted M1 macrophage polarization, activated the MEK-ERK-P65 pathway, and elevated pro-inflammatory cytokine levels. In the sclera, M2 macrophage polarization was enhanced and accompanied by increased MMP-2 expression and reduced levels of α-SMA and collagen I. Histological analysis revealed disorganized collagen fibers and widened interfibrillar spaces in the LIM sclera, indicative of active scleral remodeling. These changes were effectively counteracted by Lilrb2 overexpression, which attenuated intraocular inflammation and suppressed scleral remodeling.
Conclusion
Lilrb2 overexpression mitigates myopia progression by ameliorating intraocular inflammation and inhibiting scleral remodeling in a guinea pig model of LIM, highlighting its potential as a therapeutic target for myopia control.
{"title":"Lilrb2-mediated inhibition of scleral remodeling in experimental myopia is associated with reduced intraocular inflammation and modulation of the MEK-ERK-P65 axis","authors":"Zhongyu Ma , Jiawen Hao , Zhaohui Yang , Miao Zhang , Yuan Peng , Jizhao Xin , Yuanting Yang , Mengke Wu , Xuewei Yin , Dadong Guo , Hongsheng Bi","doi":"10.1016/j.ejphar.2025.178458","DOIUrl":"10.1016/j.ejphar.2025.178458","url":null,"abstract":"<div><h3>Objective</h3><div>This study aimed to investigate the role of leukocyte immunoglobulin-like receptor B2 (Lilrb2) in suppressing scleral remodeling in a guinea pig model of lens-induced myopia (LIM), with a focus on its ability to modulate the intraocular inflammatory microenvironment.</div></div><div><h3>Methods</h3><div>An LIM model was established in guinea pigs using −6.0 D lenses. Animals received an intravitreal injection of an adeno-associated virus overexpressing Lilrb2 (AAV-Lilrb2). Ocular biometric parameters were monitored. The activity of the MEK-ERK-P65 signaling pathway in the retina and the protein levels of MMP-2, collagen I, and α-SMA in the sclera were assessed using qPCR and Western blot. Macrophage polarization states were evaluated via multiplex immunofluorescence and Western blot analysis of iNOS (M1 marker) and ARG1 (M2 marker). Structural changes in the sclera were examined by H&E staining, while collagen organization and fibrosis were evaluated using Masson staining and immunofluorescence.</div></div><div><h3>Results</h3><div>Compared with the normal control (NC) group, the LIM group exhibited significant axial elongation and a myopic shift in refractive error. In the retina, LIM promoted M1 macrophage polarization, activated the MEK-ERK-P65 pathway, and elevated pro-inflammatory cytokine levels. In the sclera, M2 macrophage polarization was enhanced and accompanied by increased MMP-2 expression and reduced levels of α-SMA and collagen I. Histological analysis revealed disorganized collagen fibers and widened interfibrillar spaces in the LIM sclera, indicative of active scleral remodeling. These changes were effectively counteracted by Lilrb2 overexpression, which attenuated intraocular inflammation and suppressed scleral remodeling.</div></div><div><h3>Conclusion</h3><div>Lilrb2 overexpression mitigates myopia progression by ameliorating intraocular inflammation and inhibiting scleral remodeling in a guinea pig model of LIM, highlighting its potential as a therapeutic target for myopia control.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178458"},"PeriodicalIF":4.7,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145721536","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 : 2025-12-05DOI: 10.1016/j.ejphar.2025.178445
Jae-Eun Jung, Su-Bin Park, Hwa Young Yu, Su-Bin Yoon, Junghyun Kim
Xerostomia is commonly associated with aging, diabetes, and other systemic conditions. It disrupts oral homeostasis and consequently elevates the likelihood of dental caries, periodontal disorders, and oral infections. Accordingly, mitigating xerostomia is expected to help prevent these oral pathologies and improve patients’ quality of life. Because rodents cannot report subjective sensations of oral dryness, experimentally induced hyposalivation is used as an appropriate surrogate to study underlying mechanisms and potential interventions. Exendin-4 is a GLP-1 receptor agonist initially discovered in the saliva of Heloderma suspectum. It was developed as an antidiabetic medication. Using a D-galactose-based aging model in rats, this study explored the capacity of Exendin-4 to restore salivary secretion. An experimental model of hyposalivation resembling aging was created by giving rats D-galactose (300 mg/kg, intraperitoneally) each day for 7 weeks. Thereafter, Exendin-4 was provided for 2 weeks at two dosage levels (0.5 and 1 μg/head). We evaluated improvements by measuring salivary flow rates and conducting various histological and biochemical analyses. Exendin-4 treatment significantly reversed the age-related decline in salivary secretion and preserved the typical morphological characteristics of acinar cells. Histochemical staining indicated that both acidic and neutral mucin accumulation were reduced, and TUNEL analysis revealed a decrease in apoptotic cells in the salivary gland. In summary, Exendin-4 exhibits beneficial effects on D-galactose–induced hyposalivation by improving salivary secretion, preserving glandular structure, and decreasing apoptotic cell death. Thus, Exendin-4 may improve salivary secretion in animal models of aging-related hyposalivation.
{"title":"Amelioration of D-galactose-induced hyposalivation in aging rats by the GLP-1 receptor agonist Exendin-4","authors":"Jae-Eun Jung, Su-Bin Park, Hwa Young Yu, Su-Bin Yoon, Junghyun Kim","doi":"10.1016/j.ejphar.2025.178445","DOIUrl":"10.1016/j.ejphar.2025.178445","url":null,"abstract":"<div><div>Xerostomia is commonly associated with aging, diabetes, and other systemic conditions. It disrupts oral homeostasis and consequently elevates the likelihood of dental caries, periodontal disorders, and oral infections. Accordingly, mitigating xerostomia is expected to help prevent these oral pathologies and improve patients’ quality of life. Because rodents cannot report subjective sensations of oral dryness, experimentally induced hyposalivation is used as an appropriate surrogate to study underlying mechanisms and potential interventions. Exendin-4 is a GLP-1 receptor agonist initially discovered in the saliva of <em>Heloderma suspectum</em>. It was developed as an antidiabetic medication. Using a D-galactose-based aging model in rats, this study explored the capacity of Exendin-4 to restore salivary secretion. An experimental model of hyposalivation resembling aging was created by giving rats D-galactose (300 mg/kg, intraperitoneally) each day for 7 weeks. Thereafter, Exendin-4 was provided for 2 weeks at two dosage levels (0.5 and 1 μg/head). We evaluated improvements by measuring salivary flow rates and conducting various histological and biochemical analyses. Exendin-4 treatment significantly reversed the age-related decline in salivary secretion and preserved the typical morphological characteristics of acinar cells. Histochemical staining indicated that both acidic and neutral mucin accumulation were reduced, and TUNEL analysis revealed a decrease in apoptotic cells in the salivary gland. In summary, Exendin-4 exhibits beneficial effects on D-galactose–induced hyposalivation by improving salivary secretion, preserving glandular structure, and decreasing apoptotic cell death. Thus, Exendin-4 may improve salivary secretion in animal models of aging-related hyposalivation.</div></div>","PeriodicalId":12004,"journal":{"name":"European journal of pharmacology","volume":"1011 ","pages":"Article 178445"},"PeriodicalIF":4.7,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145700077","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}