Pub Date : 2025-12-01Epub Date: 2025-09-24DOI: 10.1007/s12012-025-10062-y
Rui-Qiang Qi, Juan Song, Hong-Bin Ma, Mei-Ling Nie, Liu-Hang Su, Cui-Lian Dai, Fa-Guang Zhou, Sui-Ji Li
Ventricular aneurysm is a serious complication following myocardial infarction. Increasing evidence suggests that exercise-based cardiac rehabilitation plays a protective role in cardiovascular disease. However, the effects of exercise on ventricular aneurysm and the underlying mechanisms remain poorly understood. Therefore, this study aimed to establish a murine model of ventricular aneurysm and investigate the impact of exercise on this condition, along with its potential mechanisms. In this study, using proximal coronary artery ligation, a murine cardiac ventricular aneurysm model was established and evaluated by real-time myocardial contrast echocardiography. Wild-type male C57BL/6 mice with ventricular aneurysms were randomly assigned to three groups: a Sedentary group (no exercise, n = 7), a moderate-intensity exercise group (5 m/min adaptive exercise for 2 weeks, followed by 12-m/min moderate-intensity exercise for 8 weeks, n = 9), and a high-intensity exercise group (5-m/min adaptive exercise for 2 weeks, followed by 18-m/min high-intensity exercise for 8 weeks, n = 7). After 8 weeks of exercise intervention, moderate-intensity exercise was found to significantly enhance cardiac function, reduce myocardial fibrosis, and inhibit fibroblast activation. In contrast, high-intensity exercise resulted in deteriorated cardiac function and aggravated cardiac injury. Mechanistically, this paradoxical effect was linked to the regulation of PTEN stability and subsequent modulation of Smad2/3 signaling pathway. This study provides a theoretical foundation for the role of exercise in managing ventricular aneurysms and offers insights into optimal exercise intensity levels.
{"title":"Different Intensities of Exercise Affect the Prognosis of Ventricular Aneurysm in Mice by Regulating the Progression of Cardiac Fibrosis.","authors":"Rui-Qiang Qi, Juan Song, Hong-Bin Ma, Mei-Ling Nie, Liu-Hang Su, Cui-Lian Dai, Fa-Guang Zhou, Sui-Ji Li","doi":"10.1007/s12012-025-10062-y","DOIUrl":"10.1007/s12012-025-10062-y","url":null,"abstract":"<p><p>Ventricular aneurysm is a serious complication following myocardial infarction. Increasing evidence suggests that exercise-based cardiac rehabilitation plays a protective role in cardiovascular disease. However, the effects of exercise on ventricular aneurysm and the underlying mechanisms remain poorly understood. Therefore, this study aimed to establish a murine model of ventricular aneurysm and investigate the impact of exercise on this condition, along with its potential mechanisms. In this study, using proximal coronary artery ligation, a murine cardiac ventricular aneurysm model was established and evaluated by real-time myocardial contrast echocardiography. Wild-type male C57BL/6 mice with ventricular aneurysms were randomly assigned to three groups: a Sedentary group (no exercise, n = 7), a moderate-intensity exercise group (5 m/min adaptive exercise for 2 weeks, followed by 12-m/min moderate-intensity exercise for 8 weeks, n = 9), and a high-intensity exercise group (5-m/min adaptive exercise for 2 weeks, followed by 18-m/min high-intensity exercise for 8 weeks, n = 7). After 8 weeks of exercise intervention, moderate-intensity exercise was found to significantly enhance cardiac function, reduce myocardial fibrosis, and inhibit fibroblast activation. In contrast, high-intensity exercise resulted in deteriorated cardiac function and aggravated cardiac injury. Mechanistically, this paradoxical effect was linked to the regulation of PTEN stability and subsequent modulation of Smad2/3 signaling pathway. This study provides a theoretical foundation for the role of exercise in managing ventricular aneurysms and offers insights into optimal exercise intensity levels.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1891-1902"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145129981","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 : 2025-12-01Epub Date: 2025-11-06DOI: 10.1007/s12012-025-10068-6
América Gutiérrez-Arenas, María Del Pilar Ramos-Godinez, Agustina Cano-Martínez, Francisco Correa, Angélica Ruíz-Ramírez, Elizabeth Lira-Silva, Christian Manuel Ovando Cupil, Rebeca López-Marure
Titanium dioxide (E171) is utilized in the food industry more frequently than other food additives because of its superior properties as a white colorant. Numerous in vitro studies have shown that E171 exhibits toxic effects on various cell types after a limited period of exposure. Previous observations indicate that E171 penetrates the cytoplasm of rat heart H9c2 myoblasts, leading to cell destruction after two days of treatment and impairing cardiac performance ex vivo within 10 min. Humans consume products containing E171 for extended periods; therefore, H9c2 cells were exposed to E171 once (short exposure) or multiple times (long exposure), and its toxic effects were examined after 5 weeks. Transmission electron microscopy (TEM) was used to analyze morphology and internalization. Cell counts were assessed, and cell death was evaluated using annexin plus propidium iodide (PI) and TUNEL assay. On the other hand, rats were orally administered E171 either once (acute exposure) or multiple times every other day over three months (chronic exposure), and the heart biomechanics were evaluated in a Langendorff system, as well as the infarct size by triphenyltetrazolium chloride staining. Experiments on H9c2 cells indicated that a single exposure to E171 (short) induced its absorption, a decrease in the cell number, and death after one week of exposure, and all these events were reversed after 5 weeks. Long exposure to E171 led to its internalization, inhibition of cell proliferation, alterations in cell morphology, and increased mortality during all weeks of treatment. Acute administration of E171 to rats did not induce alterations in cardiac function; however, chronic administration led to dysfunction, as indicated by a reduction in pressure and contractility index. The findings indicate that H9c2 cells and rodent hearts can recover after a brief E171 exposure, while prolonged exposure leads to substantial toxicity in both cardiac cells and cardiac tissue. Therefore, prolonged exposure to E171 could be potentially toxic to humans and lead to the development of cardiac dysfunctions.
{"title":"Effects of Short and Long Exposure to Food Additive Titanium Dioxide (E171) in H9c2 Rat Cardiomyoblasts and in the Hearts of Rats.","authors":"América Gutiérrez-Arenas, María Del Pilar Ramos-Godinez, Agustina Cano-Martínez, Francisco Correa, Angélica Ruíz-Ramírez, Elizabeth Lira-Silva, Christian Manuel Ovando Cupil, Rebeca López-Marure","doi":"10.1007/s12012-025-10068-6","DOIUrl":"10.1007/s12012-025-10068-6","url":null,"abstract":"<p><p>Titanium dioxide (E171) is utilized in the food industry more frequently than other food additives because of its superior properties as a white colorant. Numerous in vitro studies have shown that E171 exhibits toxic effects on various cell types after a limited period of exposure. Previous observations indicate that E171 penetrates the cytoplasm of rat heart H9c2 myoblasts, leading to cell destruction after two days of treatment and impairing cardiac performance ex vivo within 10 min. Humans consume products containing E171 for extended periods; therefore, H9c2 cells were exposed to E171 once (short exposure) or multiple times (long exposure), and its toxic effects were examined after 5 weeks. Transmission electron microscopy (TEM) was used to analyze morphology and internalization. Cell counts were assessed, and cell death was evaluated using annexin plus propidium iodide (PI) and TUNEL assay. On the other hand, rats were orally administered E171 either once (acute exposure) or multiple times every other day over three months (chronic exposure), and the heart biomechanics were evaluated in a Langendorff system, as well as the infarct size by triphenyltetrazolium chloride staining. Experiments on H9c2 cells indicated that a single exposure to E171 (short) induced its absorption, a decrease in the cell number, and death after one week of exposure, and all these events were reversed after 5 weeks. Long exposure to E171 led to its internalization, inhibition of cell proliferation, alterations in cell morphology, and increased mortality during all weeks of treatment. Acute administration of E171 to rats did not induce alterations in cardiac function; however, chronic administration led to dysfunction, as indicated by a reduction in pressure and contractility index. The findings indicate that H9c2 cells and rodent hearts can recover after a brief E171 exposure, while prolonged exposure leads to substantial toxicity in both cardiac cells and cardiac tissue. Therefore, prolonged exposure to E171 could be potentially toxic to humans and lead to the development of cardiac dysfunctions.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1821-1836"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145451128","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 : 2025-12-01Epub Date: 2025-11-17DOI: 10.1007/s12012-025-10071-x
Xiaomei Zhao, Kuanhang Li, Qing Liu, Miao Jiang, Yi Cao
Nanoparticle (NP) exposure has been implicated in accelerating atherosclerosis, while phytochemicals like epigallocatechin gallate (EGCG) may counteract these effects through metabolic modulation. This study investigates whether EGCG mitigates SiO2 NP-induced foam cell formation in atherosclerotic mice by restoring metabolic homeostasis. ApoE-/- mice were administered 1600 mg/kg SiO2 NPs, 80 mg/kg EGCG, or both, via daily intragastric gavage for totally 28 days. Although SiO2 NPs did not significantly alter cardiac contractile function or increase Oil Red O-positive areas in entire aortas, they markedly elevated BODIPY 493/503-positive lipid accumulation in the aortic sinus, whereas co-treatment with EGCG completely abrogated this effect. Metabolomic profiling revealed distinct perturbations induced by SiO2 NPs versus SiO2 NPs + EGCG. SiO2 NPs up-regulated amino acids (L-norvaline, leucine, N-acetylalanine) and fatty acids (decanoic acid, trans-vaccenic acid, octanoic acid) but down-regulated nucleotides (cAMP, adenine, AMP), while EGCG co-exposure reversed these changes. Pathway enrichment analysis indicated that SiO2 NPs + EGCG co-exposure more profoundly affected porphyrin metabolism (map00860) and fatty acid biosynthesis (map00061) compared to SiO2 NPs alone. Western blotting demonstrated SiO2 NP-induced activation of autophagy (increased LC3-II/I ratio) and apoptosis (increased pro-caspase 3 and cleaved caspase 3) in aortic tissue, effects reversed by EGCG. Additionally, EGCG enhanced expression of Kruppel-like factor 4 (KLF4), a key regulator of vascular homeostasis, on the lumen surface. Collectively, oral SiO2 NP exposure exacerbates early atherosclerotic progression through metabolic dysregulation and activation of autophagy-apoptosis pathways, while EGCG counteracts these effects by restoring metabolic balance and regulating KLF4.
{"title":"Oral Exposure To SiO<sub>2</sub> Nanoparticles Promotes Foam Cell Areas in Aortic Sinus of ApoE-/- Mice and Epigallocatechin Gallate Attenuates the Effects via Metabolic Restoration.","authors":"Xiaomei Zhao, Kuanhang Li, Qing Liu, Miao Jiang, Yi Cao","doi":"10.1007/s12012-025-10071-x","DOIUrl":"10.1007/s12012-025-10071-x","url":null,"abstract":"<p><p>Nanoparticle (NP) exposure has been implicated in accelerating atherosclerosis, while phytochemicals like epigallocatechin gallate (EGCG) may counteract these effects through metabolic modulation. This study investigates whether EGCG mitigates SiO<sub>2</sub> NP-induced foam cell formation in atherosclerotic mice by restoring metabolic homeostasis. ApoE-/- mice were administered 1600 mg/kg SiO<sub>2</sub> NPs, 80 mg/kg EGCG, or both, via daily intragastric gavage for totally 28 days. Although SiO<sub>2</sub> NPs did not significantly alter cardiac contractile function or increase Oil Red O-positive areas in entire aortas, they markedly elevated BODIPY 493/503-positive lipid accumulation in the aortic sinus, whereas co-treatment with EGCG completely abrogated this effect. Metabolomic profiling revealed distinct perturbations induced by SiO<sub>2</sub> NPs versus SiO<sub>2</sub> NPs + EGCG. SiO<sub>2</sub> NPs up-regulated amino acids (L-norvaline, leucine, N-acetylalanine) and fatty acids (decanoic acid, trans-vaccenic acid, octanoic acid) but down-regulated nucleotides (cAMP, adenine, AMP), while EGCG co-exposure reversed these changes. Pathway enrichment analysis indicated that SiO<sub>2</sub> NPs + EGCG co-exposure more profoundly affected porphyrin metabolism (map00860) and fatty acid biosynthesis (map00061) compared to SiO<sub>2</sub> NPs alone. Western blotting demonstrated SiO<sub>2</sub> NP-induced activation of autophagy (increased LC3-II/I ratio) and apoptosis (increased pro-caspase 3 and cleaved caspase 3) in aortic tissue, effects reversed by EGCG. Additionally, EGCG enhanced expression of Kruppel-like factor 4 (KLF4), a key regulator of vascular homeostasis, on the lumen surface. Collectively, oral SiO<sub>2</sub> NP exposure exacerbates early atherosclerotic progression through metabolic dysregulation and activation of autophagy-apoptosis pathways, while EGCG counteracts these effects by restoring metabolic balance and regulating KLF4.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1837-1849"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145534409","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 : 2025-12-01Epub Date: 2025-11-21DOI: 10.1007/s12012-025-10069-5
Alessandro Ghamlouch, Nicola Di Fazio, Maura Racciatti, Fabio Del Duca, Biancamaria Treves, Gaia De Angelis, Alessandra De Matteis, Aniello Maiese, Paola Frati
Energy drinks (EDs), widely consumed for their stimulant effects, typically contain caffeine alongside taurine, guarana, and other bioactive compounds. While generally regarded as safe, growing evidence links chronic EDs consumption to significant cardiovascular risks. Caffeine, the primary active ingredient, acts through adenosine receptor antagonism and increased calcium release, potentially provoking arrhythmias and myocardial stress. Taurine and other additives further influence cardiac excitability and contractility. This systematic review, conducted under PRISMA 2020 guidelines, investigated the cardiac histopathological consequences of chronic EDs use. A literature search spanning 2021 to March 2025 across PubMed, Google Scholar, and Scopus identified studies reporting EDs-related cardiac effects. Data extraction and analysis revealed consistent associations with QTc prolongation, atrial and ventricular arrhythmias, myocardial infarction, Takotsubo cardiomyopathy, and hypertensive episodes-even in young, healthy individuals. Animal studies support these findings, showing myocardial necrosis, myofiber disarray, mitochondrial damage, and inflammation, particularly when EDs are combined with alcohol. Notably, similarities between EDs and cocaine emerged, including shared mechanisms involving ion channel blockade, sympathetic overactivation, vasoconstriction, and prothrombotic states. Chronic use of either substance can result in structural heart damage and remodelling. Although EDs and cocaine differ in legal status and potency, their overlapping cardiovascular effects warrant greater clinical awareness and public education. Excessive EDs consumption poses a real cardiotoxic risk, especially in vulnerable populations, underscoring the need for further human research and potential regulatory consideration.
{"title":"Energy Drinks as the Legal Cocaine? A Comparative Review of Cardiac Physiopathological and Histopathological Patterns.","authors":"Alessandro Ghamlouch, Nicola Di Fazio, Maura Racciatti, Fabio Del Duca, Biancamaria Treves, Gaia De Angelis, Alessandra De Matteis, Aniello Maiese, Paola Frati","doi":"10.1007/s12012-025-10069-5","DOIUrl":"10.1007/s12012-025-10069-5","url":null,"abstract":"<p><p>Energy drinks (EDs), widely consumed for their stimulant effects, typically contain caffeine alongside taurine, guarana, and other bioactive compounds. While generally regarded as safe, growing evidence links chronic EDs consumption to significant cardiovascular risks. Caffeine, the primary active ingredient, acts through adenosine receptor antagonism and increased calcium release, potentially provoking arrhythmias and myocardial stress. Taurine and other additives further influence cardiac excitability and contractility. This systematic review, conducted under PRISMA 2020 guidelines, investigated the cardiac histopathological consequences of chronic EDs use. A literature search spanning 2021 to March 2025 across PubMed, Google Scholar, and Scopus identified studies reporting EDs-related cardiac effects. Data extraction and analysis revealed consistent associations with QTc prolongation, atrial and ventricular arrhythmias, myocardial infarction, Takotsubo cardiomyopathy, and hypertensive episodes-even in young, healthy individuals. Animal studies support these findings, showing myocardial necrosis, myofiber disarray, mitochondrial damage, and inflammation, particularly when EDs are combined with alcohol. Notably, similarities between EDs and cocaine emerged, including shared mechanisms involving ion channel blockade, sympathetic overactivation, vasoconstriction, and prothrombotic states. Chronic use of either substance can result in structural heart damage and remodelling. Although EDs and cocaine differ in legal status and potency, their overlapping cardiovascular effects warrant greater clinical awareness and public education. Excessive EDs consumption poses a real cardiotoxic risk, especially in vulnerable populations, underscoring the need for further human research and potential regulatory consideration.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1914-1926"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12662937/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145562696","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-09-23DOI: 10.1007/s12012-025-10063-x
Xiangyu Guo, Ran Wei, Ge Yang, Maoxun Huang
Heart failure (HF) is a major public health concern marked by substantial morbidity and mortality. Emerging evidences suggest that copper (Cu) may be implicated in cardiovascular diseases, but its relationship with HF remains inadequately understood. This study was conducted to investigate the association between serum copper levels and the prevalence of HF, while exploring potential underlying mechanisms using network toxicology. Data were derived from 5139 participants aged 18-80 years in the NHANES 2011-2016 cycle. Serum copper levels were measured using inductively coupled plasma dynamic reaction cell mass spectrometry (ICP-DRC-MS). Logistic regression and restricted cubic spline models were employed to evaluate the association between serum copper and HF. A network toxicology approach, incorporating database mining (CTD and ChEMBL), protein-protein interaction (PPI) network analysis, Gene Ontology (GO), and KEGG pathway enrichment, was utilized to elucidate potential molecular mechanisms. Elevated serum copper levels were significantly associated with increased prevalence of HF (OR 1.08, 95% CI 1.03-1.13; p = 0.0008). A significant sex-based interaction was observed, with a stronger association evident among males. Through in silico analysis, a total of 108 copper-related HF target genes were identified, with TP53, IL-1B, IL-6, TNF, AKT1, and ALB recognized as hub genes. Enriched KEGG pathways included lipid metabolism and atherosclerosis, apoptosis, MAPK signaling, ROS-mediated chemical carcinogenesis, and AGE-RAGE signaling in diabetic complications. Mechanistically, copper-induced oxidative stress, apoptosis, and immune dysregulation were implicated in HF pathogenesis. Serum copper levels demonstrate an independent association with HF prevalence. However, due to the cross-sectional design of the study, causal inference cannot be established. Network toxicological analysis highlights key biological pathways such as oxidative stress and apoptosis, offering mechanistic insights into the role of copper in HF development.
心力衰竭(HF)是一个主要的公共卫生问题,其特点是发病率和死亡率都很高。新出现的证据表明,铜(Cu)可能与心血管疾病有关,但其与心衰的关系仍未充分了解。本研究旨在探讨血清铜水平与心衰患病率之间的关系,同时利用网络毒理学探讨潜在的潜在机制。数据来自NHANES 2011-2016周期中5139名年龄在18-80岁的参与者。采用电感耦合血浆动态反应细胞质谱(ICP-DRC-MS)测定血清铜水平。采用Logistic回归和限制三次样条模型评价血清铜与HF的关系。利用网络毒理学方法,结合数据库挖掘(CTD和ChEMBL)、蛋白质-蛋白质相互作用(PPI)网络分析、基因本体(GO)和KEGG途径富集,来阐明潜在的分子机制。血清铜水平升高与HF患病率增加显著相关(OR 1.08, 95% CI 1.03-1.13; p = 0.0008)。我们观察到一个显著的基于性别的相互作用,在男性中有更强的关联。通过芯片分析,共鉴定出108个与铜相关的HF靶基因,其中TP53、IL-1B、IL-6、TNF、AKT1和ALB为中心基因。丰富的KEGG通路包括脂质代谢和动脉粥样硬化、细胞凋亡、MAPK信号、ros介导的化学致癌以及糖尿病并发症中的AGE-RAGE信号。在机制上,铜诱导的氧化应激、细胞凋亡和免疫失调与HF的发病有关。血清铜水平显示与心衰患病率独立相关。然而,由于研究的横断面设计,无法建立因果推理。网络毒理学分析强调了氧化应激和细胞凋亡等关键生物学途径,为铜在HF发展中的作用提供了机制见解。
{"title":"Serum Copper and Heart Failure: A Cross-Sectional and In Silico Toxicological Study.","authors":"Xiangyu Guo, Ran Wei, Ge Yang, Maoxun Huang","doi":"10.1007/s12012-025-10063-x","DOIUrl":"10.1007/s12012-025-10063-x","url":null,"abstract":"<p><p>Heart failure (HF) is a major public health concern marked by substantial morbidity and mortality. Emerging evidences suggest that copper (Cu) may be implicated in cardiovascular diseases, but its relationship with HF remains inadequately understood. This study was conducted to investigate the association between serum copper levels and the prevalence of HF, while exploring potential underlying mechanisms using network toxicology. Data were derived from 5139 participants aged 18-80 years in the NHANES 2011-2016 cycle. Serum copper levels were measured using inductively coupled plasma dynamic reaction cell mass spectrometry (ICP-DRC-MS). Logistic regression and restricted cubic spline models were employed to evaluate the association between serum copper and HF. A network toxicology approach, incorporating database mining (CTD and ChEMBL), protein-protein interaction (PPI) network analysis, Gene Ontology (GO), and KEGG pathway enrichment, was utilized to elucidate potential molecular mechanisms. Elevated serum copper levels were significantly associated with increased prevalence of HF (OR 1.08, 95% CI 1.03-1.13; p = 0.0008). A significant sex-based interaction was observed, with a stronger association evident among males. Through in silico analysis, a total of 108 copper-related HF target genes were identified, with TP53, IL-1B, IL-6, TNF, AKT1, and ALB recognized as hub genes. Enriched KEGG pathways included lipid metabolism and atherosclerosis, apoptosis, MAPK signaling, ROS-mediated chemical carcinogenesis, and AGE-RAGE signaling in diabetic complications. Mechanistically, copper-induced oxidative stress, apoptosis, and immune dysregulation were implicated in HF pathogenesis. Serum copper levels demonstrate an independent association with HF prevalence. However, due to the cross-sectional design of the study, causal inference cannot be established. Network toxicological analysis highlights key biological pathways such as oxidative stress and apoptosis, offering mechanistic insights into the role of copper in HF development.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1850-1860"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12662881/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145130011","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ferroptosis is considered to be a pathological mechanism of myocardial ischemia-reperfusion injury (MI/RI). Previous studies have shown that death-associated protein kinase 1 (DAPK1) was involved in MI/RI development, but the underlying mechanism remains unclear. Mice with ligation of the left anterior descending artery followed by reperfusion, and HL-1 cells subjected to hypoxia/reoxygenation (H/R) induction were used as MI/RI animal and cell models, respectively. The pathological status of the mouse heart was evaluated by TTC staining, HE staining and echocardiography. Cell viability was examined using CCK-8. DAPK1, SRY-box transcription factor 4 (SOX4) and ferroptosis-related indicators were determined using RT-qPCR, western blot, commercial kits and DCFH-DA method. The interaction between SOX4 and DAPK1 promoter was validated using dual luciferase assay and ChIP assay. In MI/RI mice and H/R-induced cardiomyocytes, DAPK1 and SOX4 expression was abnormally elevated compared with that in control groups. In addition, silencing of DAPK1 or SOX4 improved cardiomyocyte injury and attenuated ferroptosis in H/R-induced cardiomyocytes. At the molecular levels, SOX4 could promote DAPK1 transcription and elevate DAPK1 expression via interacting with the DAPK1 promoter. Furthermore, SOX4 knockdown alleviated cardiomyocyte injury and mitigated ferroptosis through inhibiting DAPK1 expression, thereby relieving MI/RI in mice. Our results reveled that SOX4 promoted cardiomyocyte injury and exacerbated ferroptosis, thereby intensifying MI/RI through increasing DAPK1 expression.
{"title":"SOX4 Promotes Ferroptosis to Aggravate Myocardial Ischemia/Reperfusion Injury Through Elevating DAPK1 Expression.","authors":"Lehong Yuan, Qingyu Zhao, Pengfei Yan, Jianxiong Lin, Xiangdong Qiu","doi":"10.1007/s12012-025-10072-w","DOIUrl":"10.1007/s12012-025-10072-w","url":null,"abstract":"<p><p>Ferroptosis is considered to be a pathological mechanism of myocardial ischemia-reperfusion injury (MI/RI). Previous studies have shown that death-associated protein kinase 1 (DAPK1) was involved in MI/RI development, but the underlying mechanism remains unclear. Mice with ligation of the left anterior descending artery followed by reperfusion, and HL-1 cells subjected to hypoxia/reoxygenation (H/R) induction were used as MI/RI animal and cell models, respectively. The pathological status of the mouse heart was evaluated by TTC staining, HE staining and echocardiography. Cell viability was examined using CCK-8. DAPK1, SRY-box transcription factor 4 (SOX4) and ferroptosis-related indicators were determined using RT-qPCR, western blot, commercial kits and DCFH-DA method. The interaction between SOX4 and DAPK1 promoter was validated using dual luciferase assay and ChIP assay. In MI/RI mice and H/R-induced cardiomyocytes, DAPK1 and SOX4 expression was abnormally elevated compared with that in control groups. In addition, silencing of DAPK1 or SOX4 improved cardiomyocyte injury and attenuated ferroptosis in H/R-induced cardiomyocytes. At the molecular levels, SOX4 could promote DAPK1 transcription and elevate DAPK1 expression via interacting with the DAPK1 promoter. Furthermore, SOX4 knockdown alleviated cardiomyocyte injury and mitigated ferroptosis through inhibiting DAPK1 expression, thereby relieving MI/RI in mice. Our results reveled that SOX4 promoted cardiomyocyte injury and exacerbated ferroptosis, thereby intensifying MI/RI through increasing DAPK1 expression.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1938-1951"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145534480","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}
This study aims to mine and analyze adverse events (AEs) of argatroban based on the FAERS database to better understand its safety and potential risks in the real-world. Data from the first quarter of 2004 to the third quarter of 2024 were collected, and researchers employed various signal mining methods such as Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-item Gamma Poisson Shrinker (MGPS). The study collected 54,336,884 reports of AEs, of which 2,233 were related to argatroban. Signal mining identified 110 Preferred Terms involving 25 system organ classes. There were most reports involving adults (≥ 65 years), with a slightly higher proportion in men than women. We identified known AEs, including prolonged coagulation time, hemorrhage, abnormal hepatic function. New potential AE signals were identified, such as vascular pseudoaneurysm, retroperitoneal hematoma, gangrene, thrombotic thrombocytopenic purpura, acute cardiac failure, atrial septal defect, rhabdomyolysis. The median time to event was 2.00 days, with the majority of AEs occurring within 30 days. Analysis of the FAERS database identified argatroban-associated AEs, including newly identified potential risks with the aim of enhancing clinicians' and pharmacists' awareness of drug-related risk signals and facilitate timely preventive and treatment interventions for patient safety.
{"title":"Argatroban-Associated Adverse Events: An Analysis Based on FDA Adverse Event Reporting System (FAERS).","authors":"Changyu Ren, Guimu Guo, YiXuan Wang, Tingting Chen, Jintuo Zhou, Peiguang Niu, Jinhua Zhang","doi":"10.1007/s12012-025-10066-8","DOIUrl":"10.1007/s12012-025-10066-8","url":null,"abstract":"<p><p>This study aims to mine and analyze adverse events (AEs) of argatroban based on the FAERS database to better understand its safety and potential risks in the real-world. Data from the first quarter of 2004 to the third quarter of 2024 were collected, and researchers employed various signal mining methods such as Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-item Gamma Poisson Shrinker (MGPS). The study collected 54,336,884 reports of AEs, of which 2,233 were related to argatroban. Signal mining identified 110 Preferred Terms involving 25 system organ classes. There were most reports involving adults (≥ 65 years), with a slightly higher proportion in men than women. We identified known AEs, including prolonged coagulation time, hemorrhage, abnormal hepatic function. New potential AE signals were identified, such as vascular pseudoaneurysm, retroperitoneal hematoma, gangrene, thrombotic thrombocytopenic purpura, acute cardiac failure, atrial septal defect, rhabdomyolysis. The median time to event was 2.00 days, with the majority of AEs occurring within 30 days. Analysis of the FAERS database identified argatroban-associated AEs, including newly identified potential risks with the aim of enhancing clinicians' and pharmacists' awareness of drug-related risk signals and facilitate timely preventive and treatment interventions for patient safety.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1927-1937"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312494","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 : 2025-12-01Epub Date: 2025-10-07DOI: 10.1007/s12012-025-10064-w
Qin Liu, Fenglin Xu, Ping Zhang, Xi Zheng, Fanying Xiang, Shunqing Luo, Shenying Fang, Xizhou An, Yang Bi, Jingyu Chen, Yang Gao, Shihai Zheng, Xiaohua Liang
Exposure to fine particulate matter (PM2.5) is associated with adverse cardiovascular outcomes, but the effectiveness of individual-level interventions to reduce exposure remains unclear. This meta-analysis aimed to assess the effects of four distinct categories of PM2.5 interventions on cardiovascular health including high-efficiency particulate air (HEPA) filters, respiratory protective equipment, improved cookstoves, and other behavioral PM2.5 reduction methods. We systematically searched PubMed, Embase, Web of Science, and the Cochrane library for studies published between December 2002 and March 2025 that reported the cardiovascular effect of PM2.5 reduction. Intervention methods were categorized based on their mechanisms of action and implementation settings. The primary outcomes were systolic blood pressure (SBP) and diastolic blood pressure (DBP), with secondary outcomes including endothelial function indices, heart rate variability (HRV), and inflammatory markers. A total of 72 trials involving 6,063 participants (age range: 12.4-82 years) were included. Overall, PM2.5 reduction interventions significantly decreased SBP by 1.97 mmHg (95% CI: -2.89, -1.05) and DBP by 1.08 mmHg (95% CI: -1.80, -0.35). Among secondary outcomes, C-reactive protein (CRP) decreased significantly (MD = -0.03mg/dl, 95% CI: -0.06, -0.00, P = 0.042), while no significant changes were observed in interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), or HRV. Subgroup analyses revealed differential effects by intervention type: Air filtration interventions showed the largest effect on SBP (MD = -1.94mmHg, 95% CI: -3.33, -0.55, P = 0.006) compared to other interventions. These findings suggest that PM2.5 reduction interventions could lower blood pressure and reduce markers of systemic inflammation, indicating that personal-level interventions may provide measurable cardiovascular benefits, even with short-term implementation.
{"title":"Effects of PM2.5 Reduction Interventions on Cardiovascular Indicators: A Meta-analysis.","authors":"Qin Liu, Fenglin Xu, Ping Zhang, Xi Zheng, Fanying Xiang, Shunqing Luo, Shenying Fang, Xizhou An, Yang Bi, Jingyu Chen, Yang Gao, Shihai Zheng, Xiaohua Liang","doi":"10.1007/s12012-025-10064-w","DOIUrl":"10.1007/s12012-025-10064-w","url":null,"abstract":"<p><p>Exposure to fine particulate matter (PM2.5) is associated with adverse cardiovascular outcomes, but the effectiveness of individual-level interventions to reduce exposure remains unclear. This meta-analysis aimed to assess the effects of four distinct categories of PM2.5 interventions on cardiovascular health including high-efficiency particulate air (HEPA) filters, respiratory protective equipment, improved cookstoves, and other behavioral PM2.5 reduction methods. We systematically searched PubMed, Embase, Web of Science, and the Cochrane library for studies published between December 2002 and March 2025 that reported the cardiovascular effect of PM2.5 reduction. Intervention methods were categorized based on their mechanisms of action and implementation settings. The primary outcomes were systolic blood pressure (SBP) and diastolic blood pressure (DBP), with secondary outcomes including endothelial function indices, heart rate variability (HRV), and inflammatory markers. A total of 72 trials involving 6,063 participants (age range: 12.4-82 years) were included. Overall, PM2.5 reduction interventions significantly decreased SBP by 1.97 mmHg (95% CI: -2.89, -1.05) and DBP by 1.08 mmHg (95% CI: -1.80, -0.35). Among secondary outcomes, C-reactive protein (CRP) decreased significantly (MD = -0.03mg/dl, 95% CI: -0.06, -0.00, P = 0.042), while no significant changes were observed in interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), or HRV. Subgroup analyses revealed differential effects by intervention type: Air filtration interventions showed the largest effect on SBP (MD = -1.94mmHg, 95% CI: -3.33, -0.55, P = 0.006) compared to other interventions. These findings suggest that PM2.5 reduction interventions could lower blood pressure and reduce markers of systemic inflammation, indicating that personal-level interventions may provide measurable cardiovascular benefits, even with short-term implementation.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1807-1820"},"PeriodicalIF":3.7,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145243857","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 : 2025-11-01Epub Date: 2025-09-06DOI: 10.1007/s12012-025-10058-8
Gang Deng, Yongzheng Yang, Ouyang Qing, Jiang Linhui, Su Haotao, Chi Liu, Ge Li, Moussa Ide Nasser
Myocardial infarction (MI), induced by ischemia and hypoxia of the coronary arteries, presents as myocardial necrosis. Patients often experience intense, prolonged retrosternal pain that is unrelieved by rest or nitrate therapy and is frequently associated with high blood myocardial enzyme levels. Physical effort may exacerbate this anxiety, increasing the likelihood of life-threatening consequences such as arrhythmias, shock, or cardiac failure. Chrysin, a natural flavonoid primarily found in honey and propolis, exhibits anti-inflammatory, antioxidant, anticancer, and antiviral properties. This study utilized MI models and various analytical techniques, including Western blotting, immunofluorescence, quantitative polymerase chain reaction (qPCR), and autodocking, to elucidate the molecular mechanisms underlying the action of chrysin in molecular interactions. Our results demonstrated that Chrysin alleviates apoptosis in cardiomyocytes by decreasing the Bax/Bcl-2 ratio and suppressing caspase-3 activation, actions facilitated by PPAR-γ activation and consequent overexpression of anti-apoptotic proteins. Furthermore, chrysin mitigates cardiac fibrosis by downregulating TGF-β1, collagen I, and α-SMA expression. These effects markedly diminish infarct size and improve heart function in ischemia-reperfusion damage models, ascribed to chrysin's activation of PPAR-γ and SIRT3, together with the regulation of β-catenin pathways. The preclinical data presented in this research establish a foundation for forthcoming clinical studies to assess the safety and effectiveness of chrysin in patients with myocardial infarction. This may facilitate the development of a novel treatment approach for treating MI.
{"title":"Chrysin Attenuates Myocardial Cell Apoptosis in Mice.","authors":"Gang Deng, Yongzheng Yang, Ouyang Qing, Jiang Linhui, Su Haotao, Chi Liu, Ge Li, Moussa Ide Nasser","doi":"10.1007/s12012-025-10058-8","DOIUrl":"10.1007/s12012-025-10058-8","url":null,"abstract":"<p><p>Myocardial infarction (MI), induced by ischemia and hypoxia of the coronary arteries, presents as myocardial necrosis. Patients often experience intense, prolonged retrosternal pain that is unrelieved by rest or nitrate therapy and is frequently associated with high blood myocardial enzyme levels. Physical effort may exacerbate this anxiety, increasing the likelihood of life-threatening consequences such as arrhythmias, shock, or cardiac failure. Chrysin, a natural flavonoid primarily found in honey and propolis, exhibits anti-inflammatory, antioxidant, anticancer, and antiviral properties. This study utilized MI models and various analytical techniques, including Western blotting, immunofluorescence, quantitative polymerase chain reaction (qPCR), and autodocking, to elucidate the molecular mechanisms underlying the action of chrysin in molecular interactions. Our results demonstrated that Chrysin alleviates apoptosis in cardiomyocytes by decreasing the Bax/Bcl-2 ratio and suppressing caspase-3 activation, actions facilitated by PPAR-γ activation and consequent overexpression of anti-apoptotic proteins. Furthermore, chrysin mitigates cardiac fibrosis by downregulating TGF-β1, collagen I, and α-SMA expression. These effects markedly diminish infarct size and improve heart function in ischemia-reperfusion damage models, ascribed to chrysin's activation of PPAR-γ and SIRT3, together with the regulation of β-catenin pathways. The preclinical data presented in this research establish a foundation for forthcoming clinical studies to assess the safety and effectiveness of chrysin in patients with myocardial infarction. This may facilitate the development of a novel treatment approach for treating MI.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1791-1806"},"PeriodicalIF":3.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145008117","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 : 2025-11-01Epub Date: 2025-09-20DOI: 10.1007/s12012-025-10061-z
Ling Chen, Jian Wu, Hongjie Yang, Jijun Liu, Yang Yu
Doxorubicin-induced cardiotoxicity (DIC) is pathologically characterized by oxidative stress and inflammatory cascades, creating an urgent need to identify therapeutic targets modulating these processes. While tumor necrosis factor alpha-induced protein 8 (TNFAIP8) has emerged as a regulator of inflammation and apoptosis, its functional role in DIC remains unexplored. This study systematically investigates TNFAIP8's cardioprotective mechanisms against DIC. A chronic DIC model was established in male C57BL/6 mice through intraperitoneal doxorubicin (DOX) administration (4 mg/kg weekly for 4 weeks; cumulative dose 16 mg/kg). TNFAIP8 knockdown was achieved via AAV9-delivered shRNA through tail vein injection. Multimodal assessment integrating echocardiography, histopathology analysis, and molecular profiling elucidated TNFAIP8's functional and mechanistic contributions. In DOX-induced cardiomyocytes, TNFAIP8 expression was upregulated. The absence of TNFAIP8 markedly reduced DOX-triggered oxidative stress and inflammatory responses. The potential protective mechanism of TNFAIP8 deficiency against DIC involves toll-like receptor 4 (TLR4)/NF-κB signaling pathway. Importantly, administration of the TLR4 activator lipopolysaccharide (LPS) substantially reversed the cardioprotective effects observed with TNFAIP8 deletion. Our findings establish TNFAIP8 as a critical regulator of DIC pathophysiology through TLR4/NF-κB axis modulation. Pharmacological TNFAIP8 inhibition represents a viable therapeutic strategy for mitigating chemotherapy-induced cardiac dysfunction. Future investigations should prioritize developing cardiac-targeted TNFAIP8 inhibitors and validating their efficacy in large-animal DIC models.
{"title":"TNFAIP8 Deficiency Attenuates Doxorubicin-Induced Cardiotoxicity by Inhibiting Oxidative Stress and Inflammation Via TLR4/NF-κB Signaling.","authors":"Ling Chen, Jian Wu, Hongjie Yang, Jijun Liu, Yang Yu","doi":"10.1007/s12012-025-10061-z","DOIUrl":"10.1007/s12012-025-10061-z","url":null,"abstract":"<p><p>Doxorubicin-induced cardiotoxicity (DIC) is pathologically characterized by oxidative stress and inflammatory cascades, creating an urgent need to identify therapeutic targets modulating these processes. While tumor necrosis factor alpha-induced protein 8 (TNFAIP8) has emerged as a regulator of inflammation and apoptosis, its functional role in DIC remains unexplored. This study systematically investigates TNFAIP8's cardioprotective mechanisms against DIC. A chronic DIC model was established in male C57BL/6 mice through intraperitoneal doxorubicin (DOX) administration (4 mg/kg weekly for 4 weeks; cumulative dose 16 mg/kg). TNFAIP8 knockdown was achieved via AAV9-delivered shRNA through tail vein injection. Multimodal assessment integrating echocardiography, histopathology analysis, and molecular profiling elucidated TNFAIP8's functional and mechanistic contributions. In DOX-induced cardiomyocytes, TNFAIP8 expression was upregulated. The absence of TNFAIP8 markedly reduced DOX-triggered oxidative stress and inflammatory responses. The potential protective mechanism of TNFAIP8 deficiency against DIC involves toll-like receptor 4 (TLR4)/NF-κB signaling pathway. Importantly, administration of the TLR4 activator lipopolysaccharide (LPS) substantially reversed the cardioprotective effects observed with TNFAIP8 deletion. Our findings establish TNFAIP8 as a critical regulator of DIC pathophysiology through TLR4/NF-κB axis modulation. Pharmacological TNFAIP8 inhibition represents a viable therapeutic strategy for mitigating chemotherapy-induced cardiac dysfunction. Future investigations should prioritize developing cardiac-targeted TNFAIP8 inhibitors and validating their efficacy in large-animal DIC models.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1718-1731"},"PeriodicalIF":3.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145091207","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}