Pub Date : 2025-12-22DOI: 10.1016/j.bcp.2025.117626
Manmei Li , Shuai Wu , Zhong Liu , Wei Zhang , Jing Xu , Ying Wang , Junshan Liu , Dongmei Zhang , Haiyan Tian , Yaolan Li , Wencai Ye
{"title":"Corrigendum to “Arenobufagin, a bufadienolide compound from toad venom, inhibits VEGF-mediated angiogenesis through suppression of VEGFR-2 signaling pathway” [Biochem. Pharmacol. 83(9) (2012) 1251-1260]","authors":"Manmei Li , Shuai Wu , Zhong Liu , Wei Zhang , Jing Xu , Ying Wang , Junshan Liu , Dongmei Zhang , Haiyan Tian , Yaolan Li , Wencai Ye","doi":"10.1016/j.bcp.2025.117626","DOIUrl":"10.1016/j.bcp.2025.117626","url":null,"abstract":"","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117626"},"PeriodicalIF":5.6,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145817789","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-21DOI: 10.1016/j.bcp.2025.117663
Catherine Feliu , Jean Escal , Cyril Leven , Sophie Hodin , Vanessa Rigaud , Philippe Gonzalo , Francis Coutureau , Karine Lacut , Jeremy Thereaux , Xavier Delavenne
Direct oral anticoagulants, including rivaroxaban, are widely prescribed for the prevention and treatment of thromboembolic disorders. Despite generally predictable pharmacokinetics (PK), real-world data reveal striking interindividual variability in exposure, raising concerns for both efficacy and bleeding risk. Known genetic and clinical determinants explain only part of this variability, underscoring the need to identify additional contributors. This study aimed to explore systemic protein signatures associated with rivaroxaban exposure intensity. Plasma samples were obtained from 64 participants in a phase I clinical study (NCT04180436) including healthy controls, obese patients, and post-bariatric surgery patients. Rivaroxaban PK was assessed after 20 mg once-daily dosing, and patients were stratified into high and low exposure groups. Untargeted plasma proteomic analysis was performed by liquid chromatography coupled with high-resolution mass spectrometry, followed by univariate and multivariate statistical analyses. Targeted C-reactive protein (CRP) quantification and multiplex cytokine assays were used to validate findings. Differential abundance and multivariate modeling identified inflammation and vascular related proteins associated with high rivaroxaban exposure. Notably, CRP and lipoprotein(a) (LPA) were consistently elevated in high exposure groups. Targeted CRP quantification confirmed these results, even in cases of mild to moderate inflammation. Cytokine analyses revealed a pro-inflammatory profile, characterized by increased interferon gamma and reduced Tumor Necrosis Factor alpha in high exposure patients. Protein–protein interaction networks highlighted CRP, LPA, albumin, and Apolipoprotein B as central hubs, while functional enrichment revealed pathways related to acute inflammation, oxidative stress, and vascular regulation. These findings suggest that systemic inflammation may contribute to rivaroxaban overexposure, with CRP emerging as a promising biomarker to support more personalized anticoagulant strategies.
{"title":"Inflammatory protein signatures associated with high rivaroxaban exposure","authors":"Catherine Feliu , Jean Escal , Cyril Leven , Sophie Hodin , Vanessa Rigaud , Philippe Gonzalo , Francis Coutureau , Karine Lacut , Jeremy Thereaux , Xavier Delavenne","doi":"10.1016/j.bcp.2025.117663","DOIUrl":"10.1016/j.bcp.2025.117663","url":null,"abstract":"<div><div>Direct oral anticoagulants, including rivaroxaban, are widely prescribed for the prevention and treatment of thromboembolic disorders. Despite generally predictable pharmacokinetics (PK), real-world data reveal striking interindividual variability in exposure, raising concerns for both efficacy and bleeding risk. Known genetic and clinical determinants explain only part of this variability, underscoring the need to identify additional contributors. This study aimed to explore systemic protein signatures associated with rivaroxaban exposure intensity. Plasma samples were obtained from 64 participants in a phase I clinical study (NCT04180436) including healthy controls, obese patients, and post-bariatric surgery patients. Rivaroxaban PK was assessed after 20 mg once-daily dosing, and patients were stratified into high and low exposure groups. Untargeted plasma proteomic analysis was performed by liquid chromatography coupled with high-resolution mass spectrometry, followed by univariate and multivariate statistical analyses. Targeted C-reactive protein (CRP) quantification and multiplex cytokine assays were used to validate findings. Differential abundance and multivariate modeling identified inflammation and vascular related proteins associated with high rivaroxaban exposure. Notably, CRP and lipoprotein(a) (LPA) were consistently elevated in high exposure groups. Targeted CRP quantification confirmed these results, even in cases of mild to moderate inflammation. Cytokine analyses revealed a pro-inflammatory profile, characterized by increased interferon gamma and reduced Tumor Necrosis Factor alpha in high exposure patients. Protein–protein interaction networks highlighted CRP, LPA, albumin, and Apolipoprotein B as central hubs, while functional enrichment revealed pathways related to acute inflammation, oxidative stress, and vascular regulation. These findings suggest that systemic inflammation may contribute to rivaroxaban overexposure, with CRP emerging as a promising biomarker to support more personalized anticoagulant strategies.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117663"},"PeriodicalIF":5.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145817629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
We previously found that oral administration of Panax notoginseng saponins (PNS) alleviated bone loss in ovariectomy(OVX)-induced osteoporotic mice. However, the specific active component responsible for this effect and its underlying mechanism remained unclear. Ginsenoside compound K (CK), one of the main active components of PNS, may serve as a promising therapeutic agent for osteoporosis. This study demonstrated that CK inhibited osteoclastogenesis and promoted type H vessel formation to alleviate bone loss in OVX mice. In vitro, CK concentration-dependently inhibited RANKL-induced osteoclastogenesis. In addition, high concentration CK inhibited the migration and tubule formation of HUVECs. However, treating HUVECs with CK + RANKL-stimulated RAW264.7 conditional medium showed enhancement of migration and tubule formation ability, which was blocked by adding PDGF-BB neutralising-antibody. Proteomics and network pharmacological analysis revealed CK may directly target CSF1R and inhibit osteoclast differentiation via PI3K/AKT/NFκB pathway,which subsequently conformed by drug affinity responsive target stability, cellular thermal shift, surface plasmon resonance and western blot assays. Furthermore, adding macrophage colony-stimulating factor (M-CSF) mitigated the inhibitory effect of CK on osteoclast differentiation and PI3K/AKT/NFκB pathway activation. Taken together, we demonstrated that CK exerts osteoprotective effects by targeting CSF1R to inhibit PI3K/AKT/NFκB pathway, thereby suppressing osteoclastogenesis and promoting preosteoclast PDGF-BB-induced angiogenesis.
{"title":"Ginsenoside compound K exerts osteoprotective effects by suppressing osteoclastogenesis and promoting preosteoclast PDGF-BB-induced angiogenesis via targeting CSF1R.","authors":"Shuai Chen, Bizhi Tan, Shangbin Cui, Mingbin Zhan, Yibo Wang, Fangli Huang, Tingxuan Wang, Zemin Ling, Yuan Zhang, Junnan Hu, Wei Guo, Hao Hu, Xuenong Zou","doi":"10.1016/j.bcp.2025.117661","DOIUrl":"https://doi.org/10.1016/j.bcp.2025.117661","url":null,"abstract":"<p><p>We previously found that oral administration of Panax notoginseng saponins (PNS) alleviated bone loss in ovariectomy(OVX)-induced osteoporotic mice. However, the specific active component responsible for this effect and its underlying mechanism remained unclear. Ginsenoside compound K (CK), one of the main active components of PNS, may serve as a promising therapeutic agent for osteoporosis. This study demonstrated that CK inhibited osteoclastogenesis and promoted type H vessel formation to alleviate bone loss in OVX mice. In vitro, CK concentration-dependently inhibited RANKL-induced osteoclastogenesis. In addition, high concentration CK inhibited the migration and tubule formation of HUVECs. However, treating HUVECs with CK + RANKL-stimulated RAW264.7 conditional medium showed enhancement of migration and tubule formation ability, which was blocked by adding PDGF-BB neutralising-antibody. Proteomics and network pharmacological analysis revealed CK may directly target CSF1R and inhibit osteoclast differentiation via PI3K/AKT/NFκB pathway,which subsequently conformed by drug affinity responsive target stability, cellular thermal shift, surface plasmon resonance and western blot assays. Furthermore, adding macrophage colony-stimulating factor (M-CSF) mitigated the inhibitory effect of CK on osteoclast differentiation and PI3K/AKT/NFκB pathway activation. Taken together, we demonstrated that CK exerts osteoprotective effects by targeting CSF1R to inhibit PI3K/AKT/NFκB pathway, thereby suppressing osteoclastogenesis and promoting preosteoclast PDGF-BB-induced angiogenesis.</p>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":" ","pages":"117661"},"PeriodicalIF":5.6,"publicationDate":"2025-12-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145817664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-20DOI: 10.1016/j.bcp.2025.117662
Woo Hyun Park
Inflammation represents a fundamental biological process essential for host defense and tissue integrity. However, its dysregulation, frequently characterized by a failure of active resolution, underpins a vast array of chronic pathologies. Central to this intricate process is the superfamily of eicosanoids, which are potent bioactive lipid mediators derived from 20-carbon polyunsaturated fatty acids (PUFAs). Historically, specific eicosanoids, such as prostaglandins (PGs) and leukotrienes (LTs), were regarded primarily as pro-inflammatory agents purported to be responsible for mediating vasodilation, leukocyte trafficking, nociception, and pyrexia. Consequently, therapeutic interventions like non-steroidal anti-inflammatory drugs (NSAIDs) concentrated on inhibiting their production via cyclooxygenase (COX). A significant paradigm shift posited that resolution is not passive decay but an active process orchestrated by specialized pro-resolving mediators (SPMs), including lipoxins, resolvins, protectins, and maresins. While pharmacological reports suggested these mediators direct inflammatory cessation via specific receptors, recent independent investigations have raised salient questions regarding their endogenous biosynthesis, receptor validation, and detection. This review details established biosynthetic mechanisms (COX, LOX, CYP) and contrasts classical pro-inflammatory signaling with proposed SPM actions. Crucially, a balanced perspective on the extant scientific debate is furnished, addressing the empirical challenges in reproducing SPM receptor activation—and the novel alternative mechanisms now being proposed—as well as the analytical hurdles impending their quantification. Finally, an examination is conducted regarding how a defined imbalance in these opposing mediator pathways contributes to the pathophysiology of diverse conditions, followed by a discussion of emerging therapeutic strategies that have evolved from solely inflammation-inhibition to the active promotion of its resolution.
{"title":"Eicosanoids and Inflammation: A delicate balance of Pro-Inflammatory and Pro-Resolving mediators","authors":"Woo Hyun Park","doi":"10.1016/j.bcp.2025.117662","DOIUrl":"10.1016/j.bcp.2025.117662","url":null,"abstract":"<div><div>Inflammation represents a fundamental biological process essential for host defense and tissue integrity. However, its dysregulation, frequently characterized by a failure of active resolution, underpins a vast array of chronic pathologies. Central to this intricate process is the superfamily of eicosanoids, which are potent bioactive lipid mediators derived from 20-carbon polyunsaturated fatty acids (PUFAs). Historically, specific eicosanoids, such as prostaglandins (PGs) and leukotrienes (LTs), were regarded primarily as pro-inflammatory agents purported to be responsible for mediating vasodilation, leukocyte trafficking, nociception, and pyrexia. Consequently, therapeutic interventions like non-steroidal anti-inflammatory drugs (NSAIDs) concentrated on inhibiting their production via cyclooxygenase (COX). A significant paradigm shift posited that resolution is not passive decay but an active process orchestrated by specialized pro-resolving mediators (SPMs), including lipoxins, resolvins, protectins, and maresins. While pharmacological reports suggested these mediators direct inflammatory cessation via specific receptors, recent independent investigations have raised salient questions regarding their endogenous biosynthesis, receptor validation, and detection. This review details established biosynthetic mechanisms (COX, LOX, CYP) and contrasts classical pro-inflammatory signaling with proposed SPM actions. Crucially, a balanced perspective on the extant scientific debate is furnished, addressing the empirical challenges in reproducing SPM receptor activation—and the novel alternative mechanisms now being proposed—as well as the analytical hurdles impending their quantification. Finally, an examination is conducted regarding how a defined imbalance in these opposing mediator pathways contributes to the pathophysiology of diverse conditions, followed by a discussion of emerging therapeutic strategies that have evolved from solely inflammation-inhibition to the active promotion of its resolution.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117662"},"PeriodicalIF":5.6,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145809143","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-19DOI: 10.1016/j.bcp.2025.117660
Huangxi Fu , Feng Jiang , Anqi Xu , Taicheng Zhou , Ning Liu , Xueqin Chen , Zizheng Gao , Wentong Wu , Hao Yan , Xiaochun Yang , Bo Yang , Qiaojun He , Peihua Luo , Zhifei Xu
The cardiac complications caused by drugs, including cardiac dysfunction and heart failure, significantly limit the wide clinical application of drugs and lead to morbidity and mortality. High mobility group box 1 (HMGB1) plays an extensive role in drug-induced cardiotoxicity. However, the cardiotoxic mechanisms for most small-molecule kinase inhibitors (SMKIs) remains unknown. Here, we identify that accumulated HMGB1 is associated with the cardiac complications caused by a series of FDA-approved SMKIs, among which trametinib-induced cardiomyocyte death was most significantly reversed by HMGB1 knockout. Moreover, cardiomyocyte-specific Hmgb1 deletion in mice could improve cardiac muscle contraction, calcium regulation and cardiomyocyte apoptosis in autophagy- or inflammation-independent manner. We further show that trametinib leads to the aberrant accumulation of HMGB1 by increasing its stability via inhibiting zinc finger protein Zinc Finger MYND-Type Containing 8 (ZMYND8)-mediated ubiquitination and proteasomal degradation of HMGB1, identifying ZMYND8 as a novel negative regulator of HMGB1 stability in cardiomyocyte and a potential novel regulator of cardiac function. Glycyrrhizic acid, an HMGB1 inhibitor used in clinic, prevents trametinib-induced cardiac complications. These findings reveal the mechanism and propose an effective intervention strategy for trametinib-induced cardiac complications, which would contribute to the safe application of trametinib, cardiac safety evaluation of drugs or candidate compounds and novel drug development.
{"title":"MEK inhibitor induces cardiac complications by preventing ZMYND8-mediated ubiquitination and proteasomal degradation of HMGB1","authors":"Huangxi Fu , Feng Jiang , Anqi Xu , Taicheng Zhou , Ning Liu , Xueqin Chen , Zizheng Gao , Wentong Wu , Hao Yan , Xiaochun Yang , Bo Yang , Qiaojun He , Peihua Luo , Zhifei Xu","doi":"10.1016/j.bcp.2025.117660","DOIUrl":"10.1016/j.bcp.2025.117660","url":null,"abstract":"<div><div>The cardiac complications caused by drugs, including cardiac dysfunction and heart failure, significantly limit the wide clinical application of drugs and lead to morbidity and mortality. High mobility group box 1 (HMGB1) plays an extensive role in drug-induced cardiotoxicity. However, the cardiotoxic mechanisms for most small-molecule kinase inhibitors (SMKIs) remains unknown. Here, we identify that accumulated HMGB1 is associated with the cardiac complications caused by a series of FDA-approved SMKIs, among which trametinib-induced cardiomyocyte death was most significantly reversed by <em>HMGB1</em> knockout. Moreover, cardiomyocyte-specific <em>Hmgb1</em> deletion in mice could improve cardiac muscle contraction, calcium regulation and cardiomyocyte apoptosis in autophagy- or inflammation-independent manner. We further show that trametinib leads to the aberrant accumulation of HMGB1 by increasing its stability via inhibiting zinc finger protein Zinc Finger MYND-Type Containing 8 (ZMYND8)-mediated ubiquitination and proteasomal degradation of HMGB1, identifying ZMYND8 as a novel negative regulator of HMGB1 stability in cardiomyocyte and a potential novel regulator of cardiac function. Glycyrrhizic acid, an HMGB1 inhibitor used in clinic, prevents trametinib-induced cardiac complications. These findings reveal the mechanism and propose an effective intervention strategy for trametinib-induced cardiac complications, which would contribute to the safe application of trametinib, cardiac safety evaluation of drugs or candidate compounds and novel drug development.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117660"},"PeriodicalIF":5.6,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803158","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-19DOI: 10.1016/j.bcp.2025.117659
Dan Zhang , Ju Zhu , Rui Zou , Han Zhang , Bao-Ping Yu , Pan Li , Yang Luo , Zheng Jiang , Yu Hou , Jian-Wei Zhang
Esophageal squamous cell carcinoma (ESCC) is routinely treated with platinum-based chemotherapy but almost inevitably relapses. Our previous study demonstrated that cisplatin (CDDP) induced ESCC cell senescence, and senescent cells promoted the aggressive behaviors of neighboring cancer cells through the senescence-associated secretory phenotype (SASP). Notably, the use of ‘senolytic’ drugs that selectively remove senescent cells by inducing apoptosis has been proven to improve therapeutic efficacy, but their potential application in ESCC therapy has not yet been studied. In this study, we observed that therapy-induced ESCC cell senescence was associated with poor prognosis of ESCC patients. We found that anti-apoptotic BCL-2 family member BCL-XL mediated the survival of CDDP-induced senescent ESCC cells, and senolytic drug ABT-263 (navitoclax, an inhibitor of BCL-2 and BCL-XL) selectively eliminated senescent cells by triggering apoptosis, thereby attenuating SASP-driven ESCC cell proliferation and migration in vitro and improving CDDP efficacy in a mouse model of ESCC. Mechanistically, the enhanced interaction between BCL-XL and pro-apoptotic effector protein BAX conferred apoptosis resistance in senescent ESCC cells, and ABT-263 treatment disrupted this interaction to activate apoptosis. Overall, our data indicate that CDDP-induced senescent ESCC cells could be eliminated using senolytic drugs that target BCL-XL, and thus senolytic therapy could be a potential effective strategy for improving chemotherapeutic efficacy in ESCC.
{"title":"Senolytic elimination of therapy-induced senescent cells by ABT-263 improves chemotherapeutic efficacy in esophageal squamous cell carcinoma","authors":"Dan Zhang , Ju Zhu , Rui Zou , Han Zhang , Bao-Ping Yu , Pan Li , Yang Luo , Zheng Jiang , Yu Hou , Jian-Wei Zhang","doi":"10.1016/j.bcp.2025.117659","DOIUrl":"10.1016/j.bcp.2025.117659","url":null,"abstract":"<div><div>Esophageal squamous cell carcinoma (ESCC) is routinely treated with platinum-based chemotherapy but almost inevitably relapses. Our previous study demonstrated that cisplatin (CDDP) induced ESCC cell senescence, and senescent cells promoted the aggressive behaviors of neighboring cancer cells through the senescence-associated secretory phenotype (SASP). Notably, the use of ‘senolytic’ drugs that selectively remove senescent cells by inducing apoptosis has been proven to improve therapeutic efficacy, but their potential application in ESCC therapy has not yet been studied. In this study, we observed that therapy-induced ESCC cell senescence was associated with poor prognosis of ESCC patients. We found that anti-apoptotic BCL-2 family member BCL-XL mediated the survival of CDDP-induced senescent ESCC cells, and senolytic drug ABT-263 (navitoclax, an inhibitor of BCL-2 and BCL-XL) selectively eliminated senescent cells by triggering apoptosis, thereby attenuating SASP-driven ESCC cell proliferation and migration <em>in vitro</em> and improving CDDP efficacy in a mouse model of ESCC. Mechanistically, the enhanced interaction between BCL-XL and pro-apoptotic effector protein BAX conferred apoptosis resistance in senescent ESCC cells, and ABT-263 treatment disrupted this interaction to activate apoptosis. Overall, our data indicate that CDDP-induced senescent ESCC cells could be eliminated using senolytic drugs that target BCL-XL, and thus senolytic therapy could be a potential effective strategy for improving chemotherapeutic efficacy in ESCC.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117659"},"PeriodicalIF":5.6,"publicationDate":"2025-12-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145803217","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.bcp.2025.117656
Maieryemu Waresi , Huili Zhou , Can Jiao , Jingjing Hu , Mengsha Shi , Xuanhan Lin , Tianxin Ye , Chaoyang Huang , Wei Zhang , Xiaogang Guo , Haoxuan Zhong
Arterial thrombosis is a leading cause of global cardiovascular mortality. Clinical evidence indicates that elevated IL-18 levels independently predict cardiovascular events, yet its direct role in thrombosis is poorly understood. Hence, targeting IL-18 could be a novel therapeutic strategy. This study investigated the mechanism of IL-18 in platelet activation and evaluated the antithrombotic efficacy of interleukin-18 binding protein (IL-18BP). Through in vitro and in vivo models, including Il18r1-/- mice, we demonstrated that IL-18 enhances platelet activation and thrombus formation via its receptor interleukin-18 receptor alpha (IL-18Rα). This action was mediated through the potentiation of MAPKs and PI3K/Akt signaling. Strikingly, IL-18BP exhibited superior antiplatelet effects in both mouse models and human subjects, including those with coronary artery disease (CAD). Our findings reveal that IL-18 is a key promoter of thrombosis and identify IL-18BP as a highly effective, targeted therapy for cardiovascular disease.
{"title":"IL-18 potentiates platelet activation and thrombosis through IL-18Rα-dependent MAPKs and PI3K/Akt signaling","authors":"Maieryemu Waresi , Huili Zhou , Can Jiao , Jingjing Hu , Mengsha Shi , Xuanhan Lin , Tianxin Ye , Chaoyang Huang , Wei Zhang , Xiaogang Guo , Haoxuan Zhong","doi":"10.1016/j.bcp.2025.117656","DOIUrl":"10.1016/j.bcp.2025.117656","url":null,"abstract":"<div><div>Arterial thrombosis is a leading cause of global cardiovascular mortality. Clinical evidence indicates that elevated IL-18 levels independently predict cardiovascular events, yet its direct role in thrombosis is poorly understood. Hence, targeting IL-18 could be a novel therapeutic strategy. This study investigated the mechanism of IL-18 in platelet activation and evaluated the antithrombotic efficacy of interleukin-18 binding protein (IL-18BP). Through <em>in vitro</em> and <em>in vivo</em> models, including <em>Il18r1</em><sup>-/-</sup> mice, we demonstrated that IL-18 enhances platelet activation and thrombus formation via its receptor interleukin-18 receptor alpha (IL-18Rα). This action was mediated through the potentiation of MAPKs and PI3K/Akt signaling. Strikingly, IL-18BP exhibited superior antiplatelet effects in both mouse models and human subjects, including those with coronary artery disease (CAD). Our findings reveal that IL-18 is a key promoter of thrombosis and identify IL-18BP as a highly effective, targeted therapy for cardiovascular disease.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117656"},"PeriodicalIF":5.6,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800170","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-18DOI: 10.1016/j.bcp.2025.117655
Jun Li , Jie-Chun Zhou , Wei-Ping Wen , Yu Zhou , Hui-Lin Li , Shu-Jie He , Yu-Ting Liu , Zhi-You Yang , Xiu-Hong Piao , Shu-Mei Wang , Jie Yu , Yue-Wei Ge
Ginsenoside Rg1 (Rg1), a representative saponin of ginseng, is evidenced with a remarkable neuroprotective effect; however, its direct target in neural cells has not been addressed. The present study investigated the reconstructing effect on the damaged neurite network of Rg1, and its direct target responsible for this bio-effect. The results showed that Rg1 can alleviate the Aβ25-35-induced neuronal damage and synaptic atrophy in primary neurons and PC12 cell models, respectively. Furthermore, the Septin 2, involved in cell division and cytoskeleton organization, was identified as a protein specifically binding to Rg1. In addition, the overexpression and knockdown of Septin 2 in PC12 cells supported its role in mediating the neural protective effect of Rg1. These findings uncover Septin 2 as a key target of Rg1 in neural cells, which also provides insight into the intervention mechanism of ginseng in neurological disorders.
{"title":"Septin 2: A direct target of Ginsenoside Rg1 mediating its neuroprotective effects","authors":"Jun Li , Jie-Chun Zhou , Wei-Ping Wen , Yu Zhou , Hui-Lin Li , Shu-Jie He , Yu-Ting Liu , Zhi-You Yang , Xiu-Hong Piao , Shu-Mei Wang , Jie Yu , Yue-Wei Ge","doi":"10.1016/j.bcp.2025.117655","DOIUrl":"10.1016/j.bcp.2025.117655","url":null,"abstract":"<div><div>Ginsenoside Rg1 (Rg1), a representative saponin of ginseng, is evidenced with a remarkable neuroprotective effect; however, its direct target in neural cells has not been addressed. The present study investigated the reconstructing effect on the damaged neurite network of Rg1, and its direct target responsible for this bio-effect. The results showed that Rg1 can alleviate the Aβ<sub>25-35</sub>-induced neuronal damage and synaptic atrophy in primary neurons and PC12 cell models, respectively. Furthermore, the Septin 2, involved in cell division and cytoskeleton organization, was identified as a protein specifically binding to Rg1. In addition, the overexpression and knockdown of Septin 2 in PC12 cells supported its role in mediating the neural protective effect of Rg1. These findings uncover Septin 2 as a key target of Rg1 in neural cells, which also provides insight into the intervention mechanism of ginseng in neurological disorders.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117655"},"PeriodicalIF":5.6,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1016/j.bcp.2025.117654
Hui Zhao , Zhiliang Hu
Goblet cell depletion, frequently reported as a symptom of ulcerative colitis (UC), may reflect a combination of increased mucus release, decreased mucus storage, and altered goblet cell differentiation. Here, we studied the underlying mechanisms of CCAAT/enhancer-binding protein beta (CEBPB), which is enriched in interleukin (IL)-17 signaling, in goblet cell differentiation during UC. The IL-17/CEBPB/6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) signaling axis in vivo was elicited through an IL-17A-neutralizing antibody and adeno-associated viruses targeting goblet cells. Goblet cells were used to assess the effects of IL-17/CEBPB/PFKFB3 signaling on goblet cell differentiation. IL-17 signaling activated CEBPB-mediated PFKFB3 transcription in goblet cells of dextran sodium sulfate-induced mice and drove a switch in the mode of energy metabolism from oxidative phosphorylation to aerobic glycolysis in goblet cells. The abnormal glycolytic activity of goblet cells was detrimental to cell differentiation and maturation, leading to decreased mucin secretion and weakened intestinal barrier capacity of the goblet cells, which led to the progression of UC. Overall, these results indicate that IL-17 pro-inflammatory signaling activates CEBPB/PFKFB3 expression to drive aerobic glycolysis in goblet cells, leading to goblet cell dysfunction and UC progression.
{"title":"Activation of IL-17/CEBPB/PFKFB3 triggers energy metabolic switching and goblet cell differentiation in ulcerative colitis","authors":"Hui Zhao , Zhiliang Hu","doi":"10.1016/j.bcp.2025.117654","DOIUrl":"10.1016/j.bcp.2025.117654","url":null,"abstract":"<div><div>Goblet cell depletion, frequently reported as a symptom of ulcerative colitis (UC), may reflect a combination of increased mucus release, decreased mucus storage, and altered goblet cell differentiation. Here, we studied the underlying mechanisms of CCAAT/enhancer-binding protein beta (CEBPB), which is enriched in interleukin (IL)-17 signaling, in goblet cell differentiation during UC. The IL-17/CEBPB/6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3) signaling axis <em>in vivo</em> was elicited through an IL-17A-neutralizing antibody and adeno-associated viruses targeting goblet cells. Goblet cells were used to assess the effects of IL-17/CEBPB/PFKFB3 signaling on goblet cell differentiation. IL-17 signaling activated CEBPB-mediated PFKFB3 transcription in goblet cells of dextran sodium sulfate-induced mice and drove a switch in the mode of energy metabolism from oxidative phosphorylation to aerobic glycolysis in goblet cells. The abnormal glycolytic activity of goblet cells was detrimental to cell differentiation and maturation, leading to decreased mucin secretion and weakened intestinal barrier capacity of the goblet cells, which led to the progression of UC. Overall, these results indicate that IL-17 pro-inflammatory signaling activates CEBPB/PFKFB3 expression to drive aerobic glycolysis in goblet cells, leading to goblet cell dysfunction and UC progression.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117654"},"PeriodicalIF":5.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793152","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-17DOI: 10.1016/j.bcp.2025.117658
Xiaochun Huang , Hailan Wang , Jintao Gan , Danli Zhu , Yuheng Dai , Xiaobin Wang , Li Liu , Yulin Liu , Jing Jia
Airway mucus hypersecretion is a prevalent chronic inflammatory disorder characterized by elevated mucin5AC (MUC5AC) expression, which exacerbates airflow limitation and increases susceptibility to lung infections. Transient receptor potential vanilloid 1 (TRPV1) channels, widely distributed across the airways, play a pivotal role in lung inflammation and tissue damage; however, the specific mechanism by which TRPV1 modulates airway MUC5AC expression remains unclear. To address this gap, we established an in vivo model of airway mucus hypersecretion in C57BL/6 mice via intratracheal administration of Pseudomonas aeruginosa (PA), followed by measurement of MUC5AC and TRPV1 expression levels. TRPV1 knockout mice were generated to assess lung inflammation, airway mucus secretion, and changes in MUC5AC and autophagy-related proteins following PA exposure. In vitro, an airway mucus hypersecretion model was created using Pyocyanin-treated NCI-H292 cells to measure intracellular TRPV1 and MUC5AC levels and investigate interactions between TRPV1, MUC5AC, oxidative stress, and autophagy. Both in vivo and in vitro models demonstrated that TRPV1 inhibition significantly reduced MUC5AC secretion, concurrent with decreased reactive oxygen species (ROS) production, mitigated oxidative stress, and attenuated autophagy. Further analysis revealed that the ROS scavenger N-acetylcysteine effectively decreased intracellular autophagy, while the autophagy inhibitor 3-Methyladenine significantly reduced MUC5AC secretion. Collectively, these findings indicate that TRPV1 inhibition reduces airway MUC5AC secretion through modulation of oxidative stress and autophagy pathways, suggesting TRPV1 as a promising therapeutic target for airway mucus hypersecretion diseases.
{"title":"TRPV1 mediates oxidative stress and autophagy pathways to regulate airway MUC5AC secretion","authors":"Xiaochun Huang , Hailan Wang , Jintao Gan , Danli Zhu , Yuheng Dai , Xiaobin Wang , Li Liu , Yulin Liu , Jing Jia","doi":"10.1016/j.bcp.2025.117658","DOIUrl":"10.1016/j.bcp.2025.117658","url":null,"abstract":"<div><div>Airway mucus hypersecretion is a prevalent chronic inflammatory disorder characterized by elevated mucin5AC (MUC5AC) expression, which exacerbates airflow limitation and increases susceptibility to lung infections. Transient receptor potential vanilloid 1 (TRPV1) channels, widely distributed across the airways, play a pivotal role in lung inflammation and tissue damage; however, the specific mechanism by which TRPV1 modulates airway MUC5AC expression remains unclear. To address this gap, we established an in vivo model of airway mucus hypersecretion in C57BL/6 mice via intratracheal administration of <em>Pseudomonas aeruginosa</em> (PA), followed by measurement of MUC5AC and TRPV1 expression levels. TRPV1 knockout mice were generated to assess lung inflammation, airway mucus secretion, and changes in MUC5AC and autophagy-related proteins following PA exposure. In vitro, an airway mucus hypersecretion model was created using Pyocyanin-treated NCI-H292 cells to measure intracellular TRPV1 and MUC5AC levels and investigate interactions between TRPV1, MUC5AC, oxidative stress, and autophagy. Both in vivo and <em>in vitro</em> models demonstrated that TRPV1 inhibition significantly reduced MUC5AC secretion, concurrent with decreased reactive oxygen species (ROS) production, mitigated oxidative stress, and attenuated autophagy. Further analysis revealed that the ROS scavenger N-acetylcysteine effectively decreased intracellular autophagy, while the autophagy inhibitor 3-Methyladenine significantly reduced MUC5AC secretion. Collectively, these findings indicate that TRPV1 inhibition reduces airway MUC5AC secretion through modulation of oxidative stress and autophagy pathways, suggesting TRPV1 as a promising therapeutic target for airway mucus hypersecretion diseases.</div></div>","PeriodicalId":8806,"journal":{"name":"Biochemical pharmacology","volume":"245 ","pages":"Article 117658"},"PeriodicalIF":5.6,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145793154","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}