Pub Date : 2025-11-29DOI: 10.1007/s10557-025-07816-9
Chunlong Lin, Shoushuo Fu, Huang Li, Yan Zhang, Xiaohui Li, Lun Li, Qilong Ge, Rui Li
Purpose: Pulmonary hypertension (PH) is a clinicopathological syndrome characterized by structural and functional alterations in the pulmonary vasculature arising from heterogeneous etiologies (including hypoxia) and diverse pathogenic mechanisms. These changes elevate pulmonary vascular resistance and increase pulmonary arterial pressure, ultimately progressing to right heart failure and potential fatality. Resistin-like molecule (RELM)-β activates multiple signaling pathways. This study aimed to explore the role of RELM-β in the development of chronic hypoxia-induced PH and its potential mechanisms.
Methods: Exogenous human RELM-β was injected into a mouse model of hypoxia for 3 weeks, followed by histological and hemodynamic analyses. The relationship between RELM-β and membrane proteins or receptors (OR1N1, GIPC1 and CLIC4) was determined by affinity purification-mass spectrometry (AP-MS) and co-immunoprecipitation. At the same time, in vitro cell culture experiments were carried out.
Results: Cell membrane proteins or receptors (OR1N1, GIPC1, and CLIC4) were identified as proteins interacting with RELM-β and potentially involving in the development of PH. Compared with the RELM-β overexpression group, siRNA-mediated silencing of OR1N1, GIPC1, or CLIC4 resulted in significant reduction of cell viability in both human pulmonary artery smooth muscle cells (PASMCs) and human pulmonary arterial endothelial cells (PAECs). Moreover, augmenting effect of exogenous RELM-ß on the hypoxia-induced PH was remarkably reduced in the mice with genetic deficiency of GIPC1 (GIPC1 CKO) or CLIC4 (CLIC4 CKO) compared to the wild type mice.
Conclusions: Findings of the current study suggested that RELM-β may play an important role in the development of hypoxia-induced PH through interacting with membrane proteins or receptors, including GIPC1, OR1N1, and CLIC4.
{"title":"RELM-β Augmented Hypoxia-Induced Pulmonary Hypertension Through Interacting with GIPC1, OR1N1 and CLIC4.","authors":"Chunlong Lin, Shoushuo Fu, Huang Li, Yan Zhang, Xiaohui Li, Lun Li, Qilong Ge, Rui Li","doi":"10.1007/s10557-025-07816-9","DOIUrl":"https://doi.org/10.1007/s10557-025-07816-9","url":null,"abstract":"<p><strong>Purpose: </strong>Pulmonary hypertension (PH) is a clinicopathological syndrome characterized by structural and functional alterations in the pulmonary vasculature arising from heterogeneous etiologies (including hypoxia) and diverse pathogenic mechanisms. These changes elevate pulmonary vascular resistance and increase pulmonary arterial pressure, ultimately progressing to right heart failure and potential fatality. Resistin-like molecule (RELM)-β activates multiple signaling pathways. This study aimed to explore the role of RELM-β in the development of chronic hypoxia-induced PH and its potential mechanisms.</p><p><strong>Methods: </strong>Exogenous human RELM-β was injected into a mouse model of hypoxia for 3 weeks, followed by histological and hemodynamic analyses. The relationship between RELM-β and membrane proteins or receptors (OR1N1, GIPC1 and CLIC4) was determined by affinity purification-mass spectrometry (AP-MS) and co-immunoprecipitation. At the same time, in vitro cell culture experiments were carried out.</p><p><strong>Results: </strong>Cell membrane proteins or receptors (OR1N1, GIPC1, and CLIC4) were identified as proteins interacting with RELM-β and potentially involving in the development of PH. Compared with the RELM-β overexpression group, siRNA-mediated silencing of OR1N1, GIPC1, or CLIC4 resulted in significant reduction of cell viability in both human pulmonary artery smooth muscle cells (PASMCs) and human pulmonary arterial endothelial cells (PAECs). Moreover, augmenting effect of exogenous RELM-ß on the hypoxia-induced PH was remarkably reduced in the mice with genetic deficiency of GIPC1 (GIPC1 CKO) or CLIC4 (CLIC4 CKO) compared to the wild type mice.</p><p><strong>Conclusions: </strong>Findings of the current study suggested that RELM-β may play an important role in the development of hypoxia-induced PH through interacting with membrane proteins or receptors, including GIPC1, OR1N1, and CLIC4.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145630514","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1007/s10557-025-07822-x
Eiad Habib, Ramzi Ibrahim, Hoang Nhat Pham, Mahmoud Abdelnabi, Christopher Kanaan, Abdulrhman Eldeib, Silvio Nunes Augusto, Chadi Ayoub, W H Wilson Tang, Reza Arsanjani
Purpose: Mineralocorticoid receptor antagonists (MRAs) improve outcomes in patients with heart failure with preserved ejection fraction (HFpEF), yet comparative effectiveness between steroidal and nonsteroidal agents remains unknown. Our aim was to compare cardiovascular and safety outcomes associated with finerenone versus spironolactone among patients with HFpEF.
Methods: This retrospective cohort study used a global federated health research network to identify adults with new-onset HFpEF between January 1, 2021, and August 1, 2025. Propensity score matching (1:1) balanced baseline characteristics, and outcomes were assessed with Cox proportional hazards models. A total of 253 patients prescribed finerenone and 32,266 prescribed spironolactone were identified; after matching, 251 patients were retained in each cohort (mean age, 73 years; 47% female). The primary outcome was acute HF events. Secondary outcomes included all-cause hospitalizations, mortality, myocardial infarction, stroke, and a composite of major adverse cardiovascular events. Safety outcomes included acute kidney injury (AKI) and hyperkalemia.
Results: Acute HF events occurred in 6.0% of patients receiving finerenone and 4.4% receiving spironolactone (HR 1.26, 95% CI 0.58-2.74). All-cause mortality was lower with finerenone (4.4% vs. 8.0%; HR 0.48, 95% CI 0.23-1.00). Other clinical and safety outcomes, including MACE (11.6% vs. 10.8%), AKI (10.4% vs. 8.0%), and hyperkalemia (5.8% vs. 6.2%), were similar between groups.
Conclusion: In this first direct comparison of MRAs in HFpEF, finerenone demonstrated broadly comparable effectiveness and safety to spironolactone. While these observations are hypothesis-generating, prospective studies are warranted to confirm these findings and inform therapeutic decision-making.
目的:矿化皮质激素受体拮抗剂(MRAs)改善保留射血分数(HFpEF)心力衰竭患者的预后,但甾体和非甾体药物之间的比较有效性尚不清楚。我们的目的是比较HFpEF患者中芬尼酮与螺内酯相关的心血管和安全性结果。方法:这项回顾性队列研究使用全球联邦健康研究网络来识别2021年1月1日至2025年8月1日期间新发HFpEF的成人。倾向评分匹配(1:1)平衡基线特征,并使用Cox比例风险模型评估结果。共有253例患者服用芬尼酮,32,266例服用螺内酯;匹配后,每个队列保留251例患者(平均年龄73岁,女性占47%)。主要结局是急性心衰事件。次要结局包括全因住院、死亡率、心肌梗死、卒中和主要心血管不良事件的组合。安全性结局包括急性肾损伤(AKI)和高钾血症。结果:接受芬尼酮治疗的患者中有6.0%发生急性心衰事件,接受螺内酯治疗的患者中有4.4%发生急性心衰事件(HR 1.26, 95% CI 0.58-2.74)。细芬烯酮组的全因死亡率较低(4.4%比8.0%;HR 0.48, 95% CI 0.23-1.00)。其他临床和安全性结果,包括MACE (11.6% vs. 10.8%)、AKI (10.4% vs. 8.0%)和高钾血症(5.8% vs. 6.2%),在两组之间相似。结论:在HFpEF的mra的首次直接比较中,芬烯酮显示出与螺内酯大致相当的有效性和安全性。虽然这些观察结果是假设产生的,但有必要进行前瞻性研究来证实这些发现并为治疗决策提供信息。
{"title":"Finerenone Versus Spironolactone for Heart Failure with Preserved Ejection Fraction.","authors":"Eiad Habib, Ramzi Ibrahim, Hoang Nhat Pham, Mahmoud Abdelnabi, Christopher Kanaan, Abdulrhman Eldeib, Silvio Nunes Augusto, Chadi Ayoub, W H Wilson Tang, Reza Arsanjani","doi":"10.1007/s10557-025-07822-x","DOIUrl":"https://doi.org/10.1007/s10557-025-07822-x","url":null,"abstract":"<p><strong>Purpose: </strong>Mineralocorticoid receptor antagonists (MRAs) improve outcomes in patients with heart failure with preserved ejection fraction (HFpEF), yet comparative effectiveness between steroidal and nonsteroidal agents remains unknown. Our aim was to compare cardiovascular and safety outcomes associated with finerenone versus spironolactone among patients with HFpEF.</p><p><strong>Methods: </strong>This retrospective cohort study used a global federated health research network to identify adults with new-onset HFpEF between January 1, 2021, and August 1, 2025. Propensity score matching (1:1) balanced baseline characteristics, and outcomes were assessed with Cox proportional hazards models. A total of 253 patients prescribed finerenone and 32,266 prescribed spironolactone were identified; after matching, 251 patients were retained in each cohort (mean age, 73 years; 47% female). The primary outcome was acute HF events. Secondary outcomes included all-cause hospitalizations, mortality, myocardial infarction, stroke, and a composite of major adverse cardiovascular events. Safety outcomes included acute kidney injury (AKI) and hyperkalemia.</p><p><strong>Results: </strong>Acute HF events occurred in 6.0% of patients receiving finerenone and 4.4% receiving spironolactone (HR 1.26, 95% CI 0.58-2.74). All-cause mortality was lower with finerenone (4.4% vs. 8.0%; HR 0.48, 95% CI 0.23-1.00). Other clinical and safety outcomes, including MACE (11.6% vs. 10.8%), AKI (10.4% vs. 8.0%), and hyperkalemia (5.8% vs. 6.2%), were similar between groups.</p><p><strong>Conclusion: </strong>In this first direct comparison of MRAs in HFpEF, finerenone demonstrated broadly comparable effectiveness and safety to spironolactone. While these observations are hypothesis-generating, prospective studies are warranted to confirm these findings and inform therapeutic decision-making.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145630491","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-28DOI: 10.1007/s10557-025-07818-7
Zhengzhong Wu, Kun Ke, Xin Lin, Weizhu Yang
Background: Atherosclerotic stenosis is a progressive vascular disease characterized by vascular narrowing. Bone marrow mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) have been reported to alleviate endothelial cell dysfunction.
Aim and methods: Here, we investigated the therapeutic potential of BMSC-EVs in a rabbit model of diabetic atherosclerotic stenosis and in a high-glucose-induced human umbilical vein endothelial cell (HUVEC) injury model.
Results: Histological analysis confirmed the successful establishment of the animal model. Treatment with BMSC-EVs effectively reduced intimal thickening and lipid accumulation, attenuated collagen deposition, reduced smooth muscle cell proliferation, and alleviated macrophage infiltration. Additionally, BMSC-EVs treatment decreased the expression of pro-inflammatory cytokines and improved serum lipid profiles. The HUVEC injury model recapitulates the key features of endothelial dysfunction, including decreased cell viability, increased apoptosis, oxidative stress, and proinflammatory cytokine expression. Treatment with BMSC-EVs alleviated these effects by enhancing cell viability, suppressing apoptosis, reducing oxidative stress, and lowering pro-inflammatory cytokine expression. Mechanistically, BMSC-EVs inhibited the expression of apoptotic markers, including Bax and Cleaved Caspase 3, while enhancing the phosphorylation of heat shock protein 27 (HSP27), a cytoprotective molecule, and its upstream activator P38. HSP27 knockdown reversed the anti-apoptotic and antioxidant effects of BMSC-EVs, highlighting the crucial role of HSP27 in mediating these effects.
Conclusion: Our findings suggest that BMSC-EVs hold promise as a therapeutic intervention for vascular endothelial injury associated with atherosclerosis and diabetes mellitus. Targeting HSP27 may be a potential strategy to enhance the therapeutic efficacy of BMSC-EVs in endothelial dysfunction.
{"title":"Therapeutic Potential of Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles in Atherosclerotic Stenosis and Endothelial Dysfunction.","authors":"Zhengzhong Wu, Kun Ke, Xin Lin, Weizhu Yang","doi":"10.1007/s10557-025-07818-7","DOIUrl":"https://doi.org/10.1007/s10557-025-07818-7","url":null,"abstract":"<p><strong>Background: </strong>Atherosclerotic stenosis is a progressive vascular disease characterized by vascular narrowing. Bone marrow mesenchymal stem cell-derived extracellular vesicles (BMSC-EVs) have been reported to alleviate endothelial cell dysfunction.</p><p><strong>Aim and methods: </strong>Here, we investigated the therapeutic potential of BMSC-EVs in a rabbit model of diabetic atherosclerotic stenosis and in a high-glucose-induced human umbilical vein endothelial cell (HUVEC) injury model.</p><p><strong>Results: </strong>Histological analysis confirmed the successful establishment of the animal model. Treatment with BMSC-EVs effectively reduced intimal thickening and lipid accumulation, attenuated collagen deposition, reduced smooth muscle cell proliferation, and alleviated macrophage infiltration. Additionally, BMSC-EVs treatment decreased the expression of pro-inflammatory cytokines and improved serum lipid profiles. The HUVEC injury model recapitulates the key features of endothelial dysfunction, including decreased cell viability, increased apoptosis, oxidative stress, and proinflammatory cytokine expression. Treatment with BMSC-EVs alleviated these effects by enhancing cell viability, suppressing apoptosis, reducing oxidative stress, and lowering pro-inflammatory cytokine expression. Mechanistically, BMSC-EVs inhibited the expression of apoptotic markers, including Bax and Cleaved Caspase 3, while enhancing the phosphorylation of heat shock protein 27 (HSP27), a cytoprotective molecule, and its upstream activator P38. HSP27 knockdown reversed the anti-apoptotic and antioxidant effects of BMSC-EVs, highlighting the crucial role of HSP27 in mediating these effects.</p><p><strong>Conclusion: </strong>Our findings suggest that BMSC-EVs hold promise as a therapeutic intervention for vascular endothelial injury associated with atherosclerosis and diabetes mellitus. Targeting HSP27 may be a potential strategy to enhance the therapeutic efficacy of BMSC-EVs in endothelial dysfunction.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145630507","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-25DOI: 10.1007/s10557-025-07794-y
Xinju Jia, Ting Su, Haiqiu Liang
Background: Disseminated intravascular coagulation (DIC) is a life-threatening condition characterized by uncontrolled coagulation and systemic inflammation, often triggered by sepsis. Recent studies highlight the role of N6-methyladenosine (m6A) modification in regulating endothelial dysfunction and coagulation pathways. However, the specific role of Wilms tumor 1-associated protein (WTAP)-a component of the m6A methyltransferase complex-in sepsis-induced DIC remains poorly understood.
Methods: HUVECs were stimulated with LPS to establish an in vitro inflammation model. WTAP expression was knocked down using shRNA, and its effects on intercellular adhesion molecule 1 (ICAM1) and tissue factor (TF) expression were analyzed via qPCR, Western blot, and RIP assays. The stability of ICAM1 mRNA was assessed using Actinomycin D treatment. An in vivo sepsis-induced DIC rat model was used to evaluate coagulation parameters, histopathological changes, and WTAP and ICAM1, TF expression in response to WTAP knockdown.
Results: LPS stimulation significantly increased WTAP expression and m6A modification levels in HUVECs, leading to increased ICAM1 expression and enhanced TF activity. WTAP knockdown reduced ICAM1 and TF expression, as well as TF activity. RIP and mRNA stability assays confirmed that WTAP stabilizes ICAM1 mRNA via m6A modification. In the DIC rat model, WTAP knockdown improved coagulation parameters, reduced microthrombus formation in lung and kidney tissues, and inhibited the coagulation cascade.
Conclusion: WTAP stabilizes ICAM1 mRNA via m6A modification, thereby increasing ICAM1 expression and enhancing TF activity in LPS-stimulated HUVECs. Knocking down WTAP in rats improves sepsis-induced DIC.
{"title":"Wilms Tumor 1-associated Protein Promotes Sepsis-induced Disseminated Intravascular Coagulation by Stabilizing Intercellular Adhesion Molecule 1 Via N6-methyladenosine Modification.","authors":"Xinju Jia, Ting Su, Haiqiu Liang","doi":"10.1007/s10557-025-07794-y","DOIUrl":"https://doi.org/10.1007/s10557-025-07794-y","url":null,"abstract":"<p><strong>Background: </strong>Disseminated intravascular coagulation (DIC) is a life-threatening condition characterized by uncontrolled coagulation and systemic inflammation, often triggered by sepsis. Recent studies highlight the role of N6-methyladenosine (m6A) modification in regulating endothelial dysfunction and coagulation pathways. However, the specific role of Wilms tumor 1-associated protein (WTAP)-a component of the m6A methyltransferase complex-in sepsis-induced DIC remains poorly understood.</p><p><strong>Methods: </strong>HUVECs were stimulated with LPS to establish an in vitro inflammation model. WTAP expression was knocked down using shRNA, and its effects on intercellular adhesion molecule 1 (ICAM1) and tissue factor (TF) expression were analyzed via qPCR, Western blot, and RIP assays. The stability of ICAM1 mRNA was assessed using Actinomycin D treatment. An in vivo sepsis-induced DIC rat model was used to evaluate coagulation parameters, histopathological changes, and WTAP and ICAM1, TF expression in response to WTAP knockdown.</p><p><strong>Results: </strong>LPS stimulation significantly increased WTAP expression and m6A modification levels in HUVECs, leading to increased ICAM1 expression and enhanced TF activity. WTAP knockdown reduced ICAM1 and TF expression, as well as TF activity. RIP and mRNA stability assays confirmed that WTAP stabilizes ICAM1 mRNA via m6A modification. In the DIC rat model, WTAP knockdown improved coagulation parameters, reduced microthrombus formation in lung and kidney tissues, and inhibited the coagulation cascade.</p><p><strong>Conclusion: </strong>WTAP stabilizes ICAM1 mRNA via m6A modification, thereby increasing ICAM1 expression and enhancing TF activity in LPS-stimulated HUVECs. Knocking down WTAP in rats improves sepsis-induced DIC.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145602518","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}
Purpose: this study aims to screen Protease-activated receptor 1 (PAR1) inhibitory active components with low bleeding side effects from anti-thrombin phytochemicals in Polygonum amplexicaule D. Don var. sinense Forb (PAF) to achieve multi-pathway antithrombosis without disrupting platelet hemostasis.
Methods: Docking simulation and platelet aggregation test were used to investigate interaction of PAR1-ligands to screen out anti-PAR1 active ingredients. FeCl3-induced carotid artery (CA) thrombosis and inferior vena cava (IVC)-ligated thrombosis model were constructed to collect thrombi to weigh. Histopathological analysis, coagulation function, and hemorheological indicator detection of administered rats were conducted to analyze antithrombotic effectiveness and mechanism of these compounds, which were injected in tail veins of mice to evaluate bleeding side effects.
Results: Three promising ligands (epigallocatechin gallate, homoorientin, and myricetin) with strong affinity of PAR1 were selected from PAF. Epigallocatechin gallate and homoorientin showed antithrombotic effects with decreased bleeding tendency based on activity validation, suggesting that they may be prospective and effective candidate compounds of PAR1 antagonists.
Conclusion: the integrative strategy applied to these active components allowed to rapidly discover compounds with reducing bleeding side effects during thrombus treatment, providing valuable references for precisely targeted therapy and mechanism exploration of medicinal plants.
{"title":"Protease-activated Receptor 1 Targeted Screening of Potential Antithrombotic Compounds with Hemostatic Activity from Polygonum Amplexicaule D. Don var. Sinense Forb.","authors":"Shiyi Huang, Xiangchang He, Jie Dai, Ting Zou, Hongqing Zhao, Guangming Xu","doi":"10.1007/s10557-025-07797-9","DOIUrl":"https://doi.org/10.1007/s10557-025-07797-9","url":null,"abstract":"<p><strong>Purpose: </strong>this study aims to screen Protease-activated receptor 1 (PAR1) inhibitory active components with low bleeding side effects from anti-thrombin phytochemicals in Polygonum amplexicaule D. Don var. sinense Forb (PAF) to achieve multi-pathway antithrombosis without disrupting platelet hemostasis.</p><p><strong>Methods: </strong>Docking simulation and platelet aggregation test were used to investigate interaction of PAR1-ligands to screen out anti-PAR1 active ingredients. FeCl<sub>3</sub>-induced carotid artery (CA) thrombosis and inferior vena cava (IVC)-ligated thrombosis model were constructed to collect thrombi to weigh. Histopathological analysis, coagulation function, and hemorheological indicator detection of administered rats were conducted to analyze antithrombotic effectiveness and mechanism of these compounds, which were injected in tail veins of mice to evaluate bleeding side effects.</p><p><strong>Results: </strong>Three promising ligands (epigallocatechin gallate, homoorientin, and myricetin) with strong affinity of PAR1 were selected from PAF. Epigallocatechin gallate and homoorientin showed antithrombotic effects with decreased bleeding tendency based on activity validation, suggesting that they may be prospective and effective candidate compounds of PAR1 antagonists.</p><p><strong>Conclusion: </strong>the integrative strategy applied to these active components allowed to rapidly discover compounds with reducing bleeding side effects during thrombus treatment, providing valuable references for precisely targeted therapy and mechanism exploration of medicinal plants.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145602500","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-21DOI: 10.1007/s10557-025-07813-y
Suleman Khan, Muhammad Imad Khan, Asim Shah
{"title":"Critical Evaluation of Ivabradine's Cardioprotective Potential Against Anthracycline-Induced Cardiotoxicity: A Comprehensive Review of Clinical Trials, Preclinical Studies, and Current Treatment Guidelines.","authors":"Suleman Khan, Muhammad Imad Khan, Asim Shah","doi":"10.1007/s10557-025-07813-y","DOIUrl":"https://doi.org/10.1007/s10557-025-07813-y","url":null,"abstract":"","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145562667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-20DOI: 10.1007/s10557-025-07810-1
Rami A Al-Horani, Alexandra C Selico-Dunn, Emily Lauren Schenk Smith
Purpose: Lipoprotein(a) [Lp(a)] is increasingly recognized as a genetically determined, independent risk factor for atherosclerotic cardiovascular disease (ASCVD). This review examines the structure, pathophysiology, and epidemiology of Lp(a), with a focus on its contribution to ASCVD and related conditions such as aortic valve stenosis and peripheral artery disease. The main research question addresses how Lp(a) influences cardiovascular risk and how emerging therapies may modify this risk.
Methods: This review synthesizes published evidence describing the biological characteristics of Lp(a), its mechanistic roles in disease, and its epidemiologic associations with cardiovascular outcomes. It also evaluates current and investigational therapeutic approaches by examining clinical trial data for agents targeting Lp(a).
Results: Lp(a) contributes to residual cardiovascular risk through proatherogenic, proinflammatory, and prothrombotic mechanisms. Current evidence highlights its involvement in ASCVD, aortic valve stenosis, and peripheral artery disease. Clinical studies of antisense oligonucleotides, small interfering RNAs, oral small molecules, and CRISPR-based gene editing, including pelacarsen, olpasiran, zerlasiran, lepodisiran, muvalaplin, and obicetrapib, demonstrate promising efficacy and safety. These agents show potential to significantly reduce Lp(a) levels and influence future cardiovascular prevention strategies.
Conclusion: As novel therapies advance and clinical guidelines evolve, Lp(a) is emerging as a central determinant in personalized cardiovascular care. The increasing emphasis on Lp(a) testing underscores its importance in risk stratification and future therapeutic decisionmaking.
{"title":"Lipoprotein(a) in Cardiovascular Diseases and Emerging Therapeutic Strategies.","authors":"Rami A Al-Horani, Alexandra C Selico-Dunn, Emily Lauren Schenk Smith","doi":"10.1007/s10557-025-07810-1","DOIUrl":"https://doi.org/10.1007/s10557-025-07810-1","url":null,"abstract":"<p><strong>Purpose: </strong>Lipoprotein(a) [Lp(a)] is increasingly recognized as a genetically determined, independent risk factor for atherosclerotic cardiovascular disease (ASCVD). This review examines the structure, pathophysiology, and epidemiology of Lp(a), with a focus on its contribution to ASCVD and related conditions such as aortic valve stenosis and peripheral artery disease. The main research question addresses how Lp(a) influences cardiovascular risk and how emerging therapies may modify this risk.</p><p><strong>Methods: </strong>This review synthesizes published evidence describing the biological characteristics of Lp(a), its mechanistic roles in disease, and its epidemiologic associations with cardiovascular outcomes. It also evaluates current and investigational therapeutic approaches by examining clinical trial data for agents targeting Lp(a).</p><p><strong>Results: </strong>Lp(a) contributes to residual cardiovascular risk through proatherogenic, proinflammatory, and prothrombotic mechanisms. Current evidence highlights its involvement in ASCVD, aortic valve stenosis, and peripheral artery disease. Clinical studies of antisense oligonucleotides, small interfering RNAs, oral small molecules, and CRISPR-based gene editing, including pelacarsen, olpasiran, zerlasiran, lepodisiran, muvalaplin, and obicetrapib, demonstrate promising efficacy and safety. These agents show potential to significantly reduce Lp(a) levels and influence future cardiovascular prevention strategies.</p><p><strong>Conclusion: </strong>As novel therapies advance and clinical guidelines evolve, Lp(a) is emerging as a central determinant in personalized cardiovascular care. The increasing emphasis on Lp(a) testing underscores its importance in risk stratification and future therapeutic decisionmaking.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145562643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-20DOI: 10.1007/s10557-025-07817-8
Hangli Pan, Huahong Zhang, Ying Wang, Huiying Wang
Purpose: Kawasaki disease (KD) is an acute systemic vasculitis causing coronary artery lesions (CAL). Paeoniflorin (PF) exerts anti-inflammatory and antioxidant properties, but its role in KD remains unclear. This study aims to elucidate the effects of PF on CAL and the underlying regulatory mechanisms.
Methods: C57BL/6J mice were induced by Lactobacillus casei cell wall extract (LCWE) to establish a KD mouse model. Inflammatory infiltration and pathological changes of cardiac tissue in mice was evaluated by hematoxylin eosin staining. Oxidative damage of mice was evaluated by measuring protein carbonyl, 8-hydroxy-2'-deoxyguanosine, malondialdehyde and 4-hydroxynonenal (4-HNE). Superoxide dismutase, catalase, total antioxidant capacity and glutathione peroxidase 4 was measured to evaluate antioxidant levels in mice. Inflammation levels was assessed by measuring tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6. Apoptosis of cardiac tissues was detected by TUNEL assay. The underlying mechanism was determined by RNA-sequencing.
Results: Results showed that PF inhibited inflammatory infiltration and apoptosis of cardiac tissues, as well as oxidative damage and inflammation in KD mouse model, but enhanced antioxidant levels in KD mouse model. Mechanically, PF inhibited cytochrome P450 family 2 subfamily E member 1 (CYP2E1) expression but activated NRF2 pathway in KD mouse model. CYP2E1 overexpression and ML385 (a NRF2 inhibitor) reversed the therapeutic effects of PF on KD mouse model. Moreover, CYP2E1 knockdown restored the therapeutic effects of PF inhibited by ML385 on KD mouse model.
Conclusion: Collectively, our findings demonstrated that PF alleviated CAL in KD by activating NRF2 pathway and inhibiting CYP2E1 expression. These results may provide a theoretical basis for the potential use of PF as a therapeutic agent for KD associated with CAL.
{"title":"Paeoniflorin Alleviates Coronary Artery Lesions in Kawasaki Disease by Activating NRF2 Pathway and Inhibiting CYP2E1 Expression.","authors":"Hangli Pan, Huahong Zhang, Ying Wang, Huiying Wang","doi":"10.1007/s10557-025-07817-8","DOIUrl":"https://doi.org/10.1007/s10557-025-07817-8","url":null,"abstract":"<p><strong>Purpose: </strong>Kawasaki disease (KD) is an acute systemic vasculitis causing coronary artery lesions (CAL). Paeoniflorin (PF) exerts anti-inflammatory and antioxidant properties, but its role in KD remains unclear. This study aims to elucidate the effects of PF on CAL and the underlying regulatory mechanisms.</p><p><strong>Methods: </strong>C57BL/6J mice were induced by Lactobacillus casei cell wall extract (LCWE) to establish a KD mouse model. Inflammatory infiltration and pathological changes of cardiac tissue in mice was evaluated by hematoxylin eosin staining. Oxidative damage of mice was evaluated by measuring protein carbonyl, 8-hydroxy-2'-deoxyguanosine, malondialdehyde and 4-hydroxynonenal (4-HNE). Superoxide dismutase, catalase, total antioxidant capacity and glutathione peroxidase 4 was measured to evaluate antioxidant levels in mice. Inflammation levels was assessed by measuring tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6. Apoptosis of cardiac tissues was detected by TUNEL assay. The underlying mechanism was determined by RNA-sequencing.</p><p><strong>Results: </strong>Results showed that PF inhibited inflammatory infiltration and apoptosis of cardiac tissues, as well as oxidative damage and inflammation in KD mouse model, but enhanced antioxidant levels in KD mouse model. Mechanically, PF inhibited cytochrome P450 family 2 subfamily E member 1 (CYP2E1) expression but activated NRF2 pathway in KD mouse model. CYP2E1 overexpression and ML385 (a NRF2 inhibitor) reversed the therapeutic effects of PF on KD mouse model. Moreover, CYP2E1 knockdown restored the therapeutic effects of PF inhibited by ML385 on KD mouse model.</p><p><strong>Conclusion: </strong>Collectively, our findings demonstrated that PF alleviated CAL in KD by activating NRF2 pathway and inhibiting CYP2E1 expression. These results may provide a theoretical basis for the potential use of PF as a therapeutic agent for KD associated with CAL.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145562595","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-18DOI: 10.1007/s10557-025-07798-8
Pishoy Sydhom, Bakr Al-Quraishi, Aya Gohar, Mohamad El-Shawaf, Nahla Shehata, Miral Ataya, Mark Sydhom, Nouran Awwad, Haidy Motawade, Nourhan Naji, Mahmoud Shaaban Abdelgalil
Purpose: Emerging lipid-lowering therapies, such as Plozasiran, target apolipoprotein C-III (APOC-III) by inhibiting its hepatic production at the mRNA level, presenting a novel approach to lipid regulation. However, the safety and efficacy of plozasiran have yet to be fully established.
Methods: We searched PubMed, Scopus, Web of Science, and Cochrane CENTRAL register of trials for studies comparing plozasiran to placebo in patients with dyslipidemic disorders. The primary outcomes were percentage changes from baseline in triglyceride (TG) and APOC-III levels at 24 weeks and the end of the study. Secondary outcomes included changes in other lipid parameters and safety outcomes at 24 weeks and the end of the study. A protocol was registered to PROSPERO under registration number [CRD420251026605].
Results: Four studies encompassing 1,514 participants were included in our meta-analysis. Plozasiran significantly improved TGs, APOC-III, non-high-density lipoprotein cholesterol (non-HDL-C), high-density lipoprotein cholesterol (HDL-C), and apolipoprotein B (ApoB) levels at both 24 weeks and study completion. Subgroup analyses based on dose and regimen revealed consistent findings. Quarterly administration of plozasiran at 10 mg, 25 mg, and 50 mg resulted in significant reductions in TGs, APOC-III, non-HDL-C, and HDL-C at both 24 weeks and study completion. For ApoB, all three doses produced significant reductions at 24 weeks; however, only the 25 mg and 50 mg quarterly regimens sustained these reductions through the end of the study. Regarding safety, patients receiving plozasiran experienced a higher incidence of any adverse events, headache, and mild rises in HbA1C levels. Subgroup analysis revealed a dose-dependent pattern for certain safety outcomes.
Conclusion: While Plozasiran shows strong potential as a therapeutic option for severe dyslipidemic conditions, further studies are needed to compare its efficacy and safety with currently available treatments and, more importantly, evaluate its impact on clinical outcomes for implementation in clinical practice.
目的:新兴的降脂疗法,如Plozasiran,通过在mRNA水平上抑制载脂蛋白C-III (APOC-III)的肝脏生成,提供了一种新的脂质调节方法。然而,plozasiran的安全性和有效性尚未完全确定。方法:我们检索PubMed、Scopus、Web of Science和Cochrane CENTRAL注册的试验,比较plzasiran与安慰剂在血脂异常患者中的疗效。主要结果是在24周和研究结束时甘油三酯(TG)和APOC-III水平较基线的百分比变化。次要结果包括24周和研究结束时其他脂质参数和安全性结果的变化。协议注册到PROSPERO,注册号为[CRD420251026605]。结果:我们的荟萃分析纳入了四项研究,共1,514名参与者。在24周和研究结束时,plzasiran显著改善了TGs、APOC-III、非高密度脂蛋白胆固醇(non-HDL-C)、高密度脂蛋白胆固醇(HDL-C)和载脂蛋白B (ApoB)水平。基于剂量和治疗方案的亚组分析显示了一致的结果。每季度给药10mg、25mg和50mg的plzasiran在24周和研究结束时显著降低了TGs、APOC-III、非HDL-C和HDL-C。对于ApoB,所有三种剂量在24周时均显著降低;然而,只有25毫克和50毫克的季度治疗方案在研究结束时保持了这些减少。在安全性方面,接受plzasiran治疗的患者出现了更高的不良事件发生率,头痛和HbA1C水平轻度升高。亚组分析揭示了某些安全性结果的剂量依赖模式。结论:虽然Plozasiran作为一种治疗严重血脂异常的选择具有强大的潜力,但需要进一步的研究来比较其与现有治疗方法的有效性和安全性,更重要的是,评估其在临床实践中实施对临床结果的影响。
{"title":"The Efficacy and Safety of Plozasiran on Lipid Profile in Dyslipidemic Disorders: A Systematic Review and Meta-Analysis.","authors":"Pishoy Sydhom, Bakr Al-Quraishi, Aya Gohar, Mohamad El-Shawaf, Nahla Shehata, Miral Ataya, Mark Sydhom, Nouran Awwad, Haidy Motawade, Nourhan Naji, Mahmoud Shaaban Abdelgalil","doi":"10.1007/s10557-025-07798-8","DOIUrl":"https://doi.org/10.1007/s10557-025-07798-8","url":null,"abstract":"<p><strong>Purpose: </strong>Emerging lipid-lowering therapies, such as Plozasiran, target apolipoprotein C-III (APOC-III) by inhibiting its hepatic production at the mRNA level, presenting a novel approach to lipid regulation. However, the safety and efficacy of plozasiran have yet to be fully established.</p><p><strong>Methods: </strong>We searched PubMed, Scopus, Web of Science, and Cochrane CENTRAL register of trials for studies comparing plozasiran to placebo in patients with dyslipidemic disorders. The primary outcomes were percentage changes from baseline in triglyceride (TG) and APOC-III levels at 24 weeks and the end of the study. Secondary outcomes included changes in other lipid parameters and safety outcomes at 24 weeks and the end of the study. A protocol was registered to PROSPERO under registration number [CRD420251026605].</p><p><strong>Results: </strong>Four studies encompassing 1,514 participants were included in our meta-analysis. Plozasiran significantly improved TGs, APOC-III, non-high-density lipoprotein cholesterol (non-HDL-C), high-density lipoprotein cholesterol (HDL-C), and apolipoprotein B (ApoB) levels at both 24 weeks and study completion. Subgroup analyses based on dose and regimen revealed consistent findings. Quarterly administration of plozasiran at 10 mg, 25 mg, and 50 mg resulted in significant reductions in TGs, APOC-III, non-HDL-C, and HDL-C at both 24 weeks and study completion. For ApoB, all three doses produced significant reductions at 24 weeks; however, only the 25 mg and 50 mg quarterly regimens sustained these reductions through the end of the study. Regarding safety, patients receiving plozasiran experienced a higher incidence of any adverse events, headache, and mild rises in HbA1C levels. Subgroup analysis revealed a dose-dependent pattern for certain safety outcomes.</p><p><strong>Conclusion: </strong>While Plozasiran shows strong potential as a therapeutic option for severe dyslipidemic conditions, further studies are needed to compare its efficacy and safety with currently available treatments and, more importantly, evaluate its impact on clinical outcomes for implementation in clinical practice.</p>","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145538714","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-18DOI: 10.1007/s10557-025-07815-w
Zining Chen, Yuxuan Zhang, Jian'an Wang, Jun Jiang
{"title":"Response to: Angiography‑Derived Physiology in Non‑culprit Vessels: Translational Barriers from Infarct Stratification to Pharmacotherapy Adjustment.","authors":"Zining Chen, Yuxuan Zhang, Jian'an Wang, Jun Jiang","doi":"10.1007/s10557-025-07815-w","DOIUrl":"https://doi.org/10.1007/s10557-025-07815-w","url":null,"abstract":"","PeriodicalId":9557,"journal":{"name":"Cardiovascular Drugs and Therapy","volume":" ","pages":""},"PeriodicalIF":3.1,"publicationDate":"2025-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145538509","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}