The hypothalamic paraventricular nucleus (PVN), as an important integrating center, plays a prominent role in the pathogenesis of hypertension, in maintaining the stability of cardiovascular activity through peripheral sympathetic nervous activity and secretion of various humoral factors. Acknowledging that the mechanistic targets of the endocannabinoid type 1 receptor (CB1R) are the key signaling systems involved in the regulation of hypertension, we sought to clarify whether inhibition of CB1R within the PVN ameliorates hypertension through Wnt/β-catenin/RAS pathway. Spontaneously hypertensive rats (SHRs) and Wistar Kyoto rats were randomly assigned to different groups and treated with bilateral PVN injections of AM251 (CB1R antagonist, 10 µg/h) or vehicle (artificial cerebrospinal fluid, aCSF) for four weeks. Bilateral PVN injections of AM251 significantly decreased the heart rate, the body weight and the mean arterial pressure in SHRs. AM251 lowered the expression of CB1R, Wnt3, active-β-catenin, p-IKKβ, RAS components, pro-inflammatory cytokines and elevated the expression level of Glycogen synthase kinase3β and Superoxide Dismutase in the PVN of hypertensive rats. Our findings suggest that inhibition of CB1R in the PVN ameliorates hypertension through Wnt/β-catenin/RAS pathway and broaden our current understanding of the pathological mechanism and clinical treatment of hypertension.
{"title":"Inhibition of CB1R in the Hypothalamic Paraventricular Nucleus Ameliorates Hypertension Through Wnt/β-Catenin/RAS Pathway.","authors":"Hong-Li Gao, Yu Yang, Hua Tian, Li-Yan Fu, Kai-Li Liu, Xiu-Yue Jia, Xiao-Lian Shi, Yu-Ming Kang, Xiao-Jing Yu","doi":"10.1007/s12012-024-09938-2","DOIUrl":"10.1007/s12012-024-09938-2","url":null,"abstract":"<p><p>The hypothalamic paraventricular nucleus (PVN), as an important integrating center, plays a prominent role in the pathogenesis of hypertension, in maintaining the stability of cardiovascular activity through peripheral sympathetic nervous activity and secretion of various humoral factors. Acknowledging that the mechanistic targets of the endocannabinoid type 1 receptor (CB1R) are the key signaling systems involved in the regulation of hypertension, we sought to clarify whether inhibition of CB1R within the PVN ameliorates hypertension through Wnt/β-catenin/RAS pathway. Spontaneously hypertensive rats (SHRs) and Wistar Kyoto rats were randomly assigned to different groups and treated with bilateral PVN injections of AM251 (CB1R antagonist, 10 µg/h) or vehicle (artificial cerebrospinal fluid, aCSF) for four weeks. Bilateral PVN injections of AM251 significantly decreased the heart rate, the body weight and the mean arterial pressure in SHRs. AM251 lowered the expression of CB1R, Wnt3, active-β-catenin, p-IKKβ, RAS components, pro-inflammatory cytokines and elevated the expression level of Glycogen synthase kinase3β and Superoxide Dismutase in the PVN of hypertensive rats. Our findings suggest that inhibition of CB1R in the PVN ameliorates hypertension through Wnt/β-catenin/RAS pathway and broaden our current understanding of the pathological mechanism and clinical treatment of hypertension.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"9-23"},"PeriodicalIF":3.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521093","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 : 2025-01-01Epub Date: 2024-12-18DOI: 10.1007/s12012-024-09948-0
Muhammad Fahad Tahir, Xiaofei Wu, Yuwei Wang, Qin Liu, Xizhou An, Daochao Huang, Lijing Chen, Lanling Chen, Xiaohua Liang
A limited number of cohort studies have explored the impact of serum essential metal elements on blood pressure (BP) or glycolipids and their regulatory mechanism in children. This study aimed to investigate the relationship between serum metal concentrations of iron (Fe), zinc (Zn), calcium (Ca), copper (Cu), and magnesium (Mg) and BP in children, and explore the potential mediating effects of glycolipid profiles. This cohort study included 1993 children (3566 BP measurements) aged 6-14 years in Chongqing, China. Serum essential metals, BP, lipid profiles, and glucose and insulin levels were measured. The relationship between serum metal levels and BP was analyzed using generalized linear and regression models, and a mediation analysis was performed to examine the potential mediating role of glycolipids. After adjusting for confounders, positive associations were found between serum Fe and Zn levels and BP parameters (all P < 0.05). A "U" style relationship between Cu and BP was found. Stronger associations were found in children aged ≤ 10 years, with sex-specific differences for Fe, Zn, and Cu. The relationship between elevated BP and serum Mg and Ca was not found. Our study found that triglycerides showed a significant relationship with Fe and Zn levels (P < 0.005). Moreover, triglycerides, partially mediate the effects of Zn on elevated BP. Serum Fe, Zn, and Cu concentrations were associated with BP in children, and age and sex differences were observed. Triglycerides may play a mediating role. These findings highlight the importance of maintaining an optimal serum essential metal status for cardiovascular health in children and suggest potential early prevention strategies.
{"title":"Association Between Serum Essential Metal Elements and Blood Pressure in Children: A Cohort Study.","authors":"Muhammad Fahad Tahir, Xiaofei Wu, Yuwei Wang, Qin Liu, Xizhou An, Daochao Huang, Lijing Chen, Lanling Chen, Xiaohua Liang","doi":"10.1007/s12012-024-09948-0","DOIUrl":"10.1007/s12012-024-09948-0","url":null,"abstract":"<p><p>A limited number of cohort studies have explored the impact of serum essential metal elements on blood pressure (BP) or glycolipids and their regulatory mechanism in children. This study aimed to investigate the relationship between serum metal concentrations of iron (Fe), zinc (Zn), calcium (Ca), copper (Cu), and magnesium (Mg) and BP in children, and explore the potential mediating effects of glycolipid profiles. This cohort study included 1993 children (3566 BP measurements) aged 6-14 years in Chongqing, China. Serum essential metals, BP, lipid profiles, and glucose and insulin levels were measured. The relationship between serum metal levels and BP was analyzed using generalized linear and regression models, and a mediation analysis was performed to examine the potential mediating role of glycolipids. After adjusting for confounders, positive associations were found between serum Fe and Zn levels and BP parameters (all P < 0.05). A \"U\" style relationship between Cu and BP was found. Stronger associations were found in children aged ≤ 10 years, with sex-specific differences for Fe, Zn, and Cu. The relationship between elevated BP and serum Mg and Ca was not found. Our study found that triglycerides showed a significant relationship with Fe and Zn levels (P < 0.005). Moreover, triglycerides, partially mediate the effects of Zn on elevated BP. Serum Fe, Zn, and Cu concentrations were associated with BP in children, and age and sex differences were observed. Triglycerides may play a mediating role. These findings highlight the importance of maintaining an optimal serum essential metal status for cardiovascular health in children and suggest potential early prevention strategies.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"121-134"},"PeriodicalIF":3.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142846145","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}
Histone demethylation in cardiac hypertrophy is poorly understood. This study aims to determine the role of the histone demethylase LSD1 in pathological cardiac hypertrophy. Both isoprenaline (ISO)-treated and transverse aortic constriction (TAC)-treated rats developed hypertrophic hearts. LSD1 was significantly decreased; the histone marks mono- and dimethyl H3K4 and H3K9 (H3K4me1/2 and H3K9me1/2) were significantly up-regulated in the hypertrophic heart tissue, as well as the expression of the ANP, α-HMC and MLV-2v genes. An LSD1 inhibitor, OG-L002 could also induce cardiac hypertrophy and enhance the induction of cardiac hypertrophy by ISO. Overexpressed LSD1 abolished ISO-induced cardiac hypertrophy and downregulated H3K4me1/2 and H3K9me1/2 expression. Overexpression of LSD1 also reduced the expression of ANP, α-HMC and MLV-2v. In addition, we have reported isoprenaline (ISO) as one of the histone demethylase LSD1 inhibitors. This was confirmed by molecular docking, molecular dynamic studies and a histone demethylation assay. The H3K4me1/2 expression increases with the incubation of ISO in HEK 293T and HELA cells. CaMKII could be significantly activated by the LSD1 inhibitor OG-L002 as well as by ISO in rats. In summary, we have identified a novel role for LSD1 in initiating and maintaining cardiac hypertrophy.
人们对组蛋白去甲基化在心肌肥大中的作用知之甚少。本研究旨在确定组蛋白去甲基化酶 LSD1 在病理性心肌肥厚中的作用。异丙肾上腺素(ISO)处理过的大鼠和横主动脉收缩(TAC)处理过的大鼠都出现了肥厚性心脏。肥大的心脏组织中,LSD1明显减少;组蛋白标记单二甲基H3K4和H3K9(H3K4me1/2和H3K9me1/2)明显上调,ANP、α-HMC和MLV-2v基因的表达也明显上调。LSD1抑制剂OG-L002也能诱导心脏肥大,并增强ISO对心脏肥大的诱导作用。过表达LSD1可消除ISO诱导的心肌肥大,并下调H3K4me1/2和H3K9me1/2的表达。过表达 LSD1 还会降低 ANP、α-HMC 和 MLV-2v 的表达。此外,我们还发现异丙肾上腺素(ISO)是组蛋白去甲基化酶 LSD1 的抑制剂之一。分子对接、分子动力学研究和组蛋白去甲基化试验证实了这一点。在 HEK 293T 和 HELA 细胞中,H3K4me1/2 的表达随着 ISO 的孵育而增加。在大鼠体内,LSD1 抑制剂 OG-L002 和 ISO 均可显著激活 CaMKII。总之,我们发现了 LSD1 在启动和维持心肌肥大中的新作用。
{"title":"Isoprenaline Inhibits Histone Demethylase LSD1 to Induce Cardiac Hypertrophy.","authors":"Lili Wu, Bo Yang, Yingze Sun, Guanwei Fan, Lina Ma, Ying Ma, Xianjia Xiong, Hui Zhou, Huiping Wang, Ling Zhang, Bing Yang","doi":"10.1007/s12012-024-09937-3","DOIUrl":"10.1007/s12012-024-09937-3","url":null,"abstract":"<p><p>Histone demethylation in cardiac hypertrophy is poorly understood. This study aims to determine the role of the histone demethylase LSD1 in pathological cardiac hypertrophy. Both isoprenaline (ISO)-treated and transverse aortic constriction (TAC)-treated rats developed hypertrophic hearts. LSD1 was significantly decreased; the histone marks mono- and dimethyl H3K4 and H3K9 (H3K4me1/2 and H3K9me1/2) were significantly up-regulated in the hypertrophic heart tissue, as well as the expression of the ANP, α-HMC and MLV-2v genes. An LSD1 inhibitor, OG-L002 could also induce cardiac hypertrophy and enhance the induction of cardiac hypertrophy by ISO. Overexpressed LSD1 abolished ISO-induced cardiac hypertrophy and downregulated H3K4me1/2 and H3K9me1/2 expression. Overexpression of LSD1 also reduced the expression of ANP, α-HMC and MLV-2v. In addition, we have reported isoprenaline (ISO) as one of the histone demethylase LSD1 inhibitors. This was confirmed by molecular docking, molecular dynamic studies and a histone demethylation assay. The H3K4me1/2 expression increases with the incubation of ISO in HEK 293T and HELA cells. CaMKII could be significantly activated by the LSD1 inhibitor OG-L002 as well as by ISO in rats. In summary, we have identified a novel role for LSD1 in initiating and maintaining cardiac hypertrophy.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"34-47"},"PeriodicalIF":3.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615325","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}
Diabetic cardiomyopathy (DCM) is a common and severe complication of Diabetes mellitus (DM). Dapagliflozin (DAPA) is an oral anti-diabetic drug worldwide for the treatment of type 2 DM. However, the action and mechanism of DAPA in cardiac fibrosis during DCM remain vague. Primary cardiac fibroblasts (CFs) were incubated with high glucose (HG) in vitro. Cell proliferation was detected by MTT and EdU assays. Oxidative stress was evaluated by determining the production of reactive oxygen species and malondialdehyde. Cell fibrosis was assessed by detecting fibrosis-related proteins by western blotting. Levels of Mettl3 (Methyltransferase 3) and Marcks (myristoylated alanine-rich C kinase substrate) were measured using qRT-PCR and western blotting. The m6A modification profile was determined by methylated RNA immunoprecipitation assay and the interaction between Mettl3 and Marcks was verified using dual-luciferase reporter and RIP assays. DAPA treatment alleviated HG-induced proliferation, oxidative stress, and fibrosis in CFs. HG promoted the expression of Mettl3 in CFs. Knockdown of Mettl3 reversed HG-induced proliferation, oxidative stress, and fibrosis in CFs; moreover, forced expression of Mettl3 abolished the protective effects of DAPA on CFs under HG condition. Mechanistically, Mettl3 interacted with Marcks in CFs and induced Marcks mRNA m6A modification. HG induced high expression of Marcks in CFs. The overexpression of Marcks could counteract DAPA or Mettl3 knockdown-evoked inhibitory effects on CF proliferation, oxidative stress, and fibrosis under HG condition. Dapagliflozin suppressed HG-induced proliferation, oxidative stress, and fibrosis by reducing Mettl3-induced m6A modification in Marcks mRNA.
糖尿病心肌病(DCM)是糖尿病(DM)常见的严重并发症。达帕格列净(DAPA)是一种全球通用的口服抗糖尿病药物,用于治疗2型糖尿病。然而,DAPA对DCM期间心脏纤维化的作用和机制仍然模糊不清。原代心脏成纤维细胞(CFs)与高葡萄糖(HG)体外培养。通过 MTT 和 EdU 试验检测细胞增殖。通过测定活性氧和丙二醛的产生来评估氧化应激。通过 Western 印迹法检测纤维化相关蛋白,评估细胞纤维化情况。采用 qRT-PCR 和 Western 印迹法测定了 Mettl3(甲基转移酶 3)和 Marcks(肉豆蔻酰化富丙氨酸 C 激酶底物)的水平。通过甲基化 RNA 免疫沉淀分析确定了 m6A 修饰概况,并通过双荧光素酶报告和 RIP 分析验证了 Mettl3 和 Marcks 之间的相互作用。DAPA处理减轻了HG诱导的CFs增殖、氧化应激和纤维化。HG促进了Mettl3在CFs中的表达。敲除Mettl3可逆转HG诱导的CFs增殖、氧化应激和纤维化;此外,在HG条件下,强制表达Mettl3可取消DAPA对CFs的保护作用。从机制上看,Mettl3与CFs中的Marcks相互作用,诱导Marcks mRNA m6A修饰。HG 诱导了 Marcks 在 CFs 中的高表达。在HG条件下,Marcks的过表达可以抵消DAPA或Mettl3基因敲除对CF增殖、氧化应激和纤维化的抑制作用。达帕格列净通过减少Mettl3诱导的Marcks mRNA m6A修饰,抑制了HG诱导的增殖、氧化应激和纤维化。
{"title":"Dapagliflozin Suppresses High Glucose-Induced Proliferation, Oxidative Stress, and Fibrosis by Reducing Mettl3-Induced m6A Modification in Marcks mRNA.","authors":"Binhao Shi, Jianfei Wang, Jing Zhang, Ji Li, Yancheng Hao, Xianhe Lin, Ren Zhao","doi":"10.1007/s12012-024-09945-3","DOIUrl":"10.1007/s12012-024-09945-3","url":null,"abstract":"<p><p>Diabetic cardiomyopathy (DCM) is a common and severe complication of Diabetes mellitus (DM). Dapagliflozin (DAPA) is an oral anti-diabetic drug worldwide for the treatment of type 2 DM. However, the action and mechanism of DAPA in cardiac fibrosis during DCM remain vague. Primary cardiac fibroblasts (CFs) were incubated with high glucose (HG) in vitro. Cell proliferation was detected by MTT and EdU assays. Oxidative stress was evaluated by determining the production of reactive oxygen species and malondialdehyde. Cell fibrosis was assessed by detecting fibrosis-related proteins by western blotting. Levels of Mettl3 (Methyltransferase 3) and Marcks (myristoylated alanine-rich C kinase substrate) were measured using qRT-PCR and western blotting. The m6A modification profile was determined by methylated RNA immunoprecipitation assay and the interaction between Mettl3 and Marcks was verified using dual-luciferase reporter and RIP assays. DAPA treatment alleviated HG-induced proliferation, oxidative stress, and fibrosis in CFs. HG promoted the expression of Mettl3 in CFs. Knockdown of Mettl3 reversed HG-induced proliferation, oxidative stress, and fibrosis in CFs; moreover, forced expression of Mettl3 abolished the protective effects of DAPA on CFs under HG condition. Mechanistically, Mettl3 interacted with Marcks in CFs and induced Marcks mRNA m6A modification. HG induced high expression of Marcks in CFs. The overexpression of Marcks could counteract DAPA or Mettl3 knockdown-evoked inhibitory effects on CF proliferation, oxidative stress, and fibrosis under HG condition. Dapagliflozin suppressed HG-induced proliferation, oxidative stress, and fibrosis by reducing Mettl3-induced m6A modification in Marcks mRNA.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"110-120"},"PeriodicalIF":3.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142666972","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}
Congenital heart disease (CHD) is a major cause of infant mortality and morbidity, with growing interest in the role of environmental factors in its etiology. Di-(2-ethylhexyl) phthalate (DEHP), an environmental endocrine disruptor, has been implicated in the development of CHD. This study aimed to investigate the effects of DEHP exposure on fetal heart development in mice. Pregnant mice exposed to DEHP exhibited increased fetal malformations, decreased fetal weight, and reduced crown-rump length.f Transcriptomic analysis revealed the downregulation of genes involved in aerobic respiration and mitochondrial ATP synthesis. Functional assays demonstrated reduced mitochondrial respiration, decreased ATP production, elevated reactive oxygen species levels, and lowered mitochondrial membrane potential in DEHP-exposed fetal cardiomyocytes. These findings underscore the detrimental effects of DEHP on fetal cardiac health and provide insights into the molecular mechanisms underlying DEHP-induced CHD. Understanding these mechanisms is crucial for developing preventive strategies against environmental toxicants that affect fetal cardiac development.
先天性心脏病(CHD)是婴儿死亡和发病的主要原因之一,人们越来越关注环境因素在其病因中的作用。邻苯二甲酸二(2-乙基己基)酯(DEHP)是一种环境内分泌干扰物,被认为与先天性心脏病的发病有关。本研究旨在调查接触 DEHP 对小鼠胎儿心脏发育的影响。转录组分析显示,参与有氧呼吸和线粒体 ATP 合成的基因下调。功能测试显示,在暴露于 DEHP 的胎儿心肌细胞中,线粒体呼吸减少,ATP 生成减少,活性氧水平升高,线粒体膜电位降低。这些发现强调了DEHP对胎儿心脏健康的有害影响,并提供了对DEHP诱发先天性心脏病的分子机制的见解。了解这些机制对于制定针对影响胎儿心脏发育的环境毒物的预防策略至关重要。
{"title":"Di-(2-ethylhexyl) Phthalate Exposure Induces Developmental Toxicity in the Mouse Fetal Heart via Mitochondrial Dysfunction.","authors":"Yafei Guo, Bowen Li, Yu Yan, Nanjun Zhang, Shuran Shao, Lixia Yang, Lixue Ouyang, Ping Wu, Fan Ma, Hongyu Duan, Kaiyu Zhou, Yimin Hua, Chuan Wang","doi":"10.1007/s12012-024-09936-4","DOIUrl":"10.1007/s12012-024-09936-4","url":null,"abstract":"<p><p>Congenital heart disease (CHD) is a major cause of infant mortality and morbidity, with growing interest in the role of environmental factors in its etiology. Di-(2-ethylhexyl) phthalate (DEHP), an environmental endocrine disruptor, has been implicated in the development of CHD. This study aimed to investigate the effects of DEHP exposure on fetal heart development in mice. Pregnant mice exposed to DEHP exhibited increased fetal malformations, decreased fetal weight, and reduced crown-rump length.f Transcriptomic analysis revealed the downregulation of genes involved in aerobic respiration and mitochondrial ATP synthesis. Functional assays demonstrated reduced mitochondrial respiration, decreased ATP production, elevated reactive oxygen species levels, and lowered mitochondrial membrane potential in DEHP-exposed fetal cardiomyocytes. These findings underscore the detrimental effects of DEHP on fetal cardiac health and provide insights into the molecular mechanisms underlying DEHP-induced CHD. Understanding these mechanisms is crucial for developing preventive strategies against environmental toxicants that affect fetal cardiac development.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"48-57"},"PeriodicalIF":3.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142495720","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 : 2025-01-01Epub Date: 2024-11-11DOI: 10.1007/s12012-024-09944-4
Maria Fernanda Conte Bernhardt, Nathália Ronconi-Krüger, Evelise Maria Nazari
In recent years, concerns have been raised regarding the safety of exposure to pyriproxyfen (PPF), a larvicide commonly used in drinking water reservoirs to control populations of disease-vector mosquitoes for human safety. These concerns are focused mainly on exposure by pregnant women, since studies have shown deleterious effects of PPF on embryonic development, mainly addressing the central nervous system. However, since previous studies showed reduced growth in embryos exposed to PPF, we hypothesize that PPF exposure impairs the cardiovascular system, responsible for ensuring appropriate blood supply, which leads to stunted growth. This study aimed to investigate the impact of PPF exposure on heart ventricular morphology, its influence on cell proliferation and apoptosis, as well as assess the impact on the functionality of the heart and on embryonic growth. Chicken embryos were used as a model and two sublethal concentrations were tested: 0.01 mg/L and 10 mg/L PPF. Thinning of cardiac tissue was evident in heart structures at 10 mg/L PPF. Furthermore, DNA double-strand breaks and reduced cell proliferation were observed, combined with decreased apoptosis suggesting cell cycle arrest, especially in the left ventricle for both concentrations. In addition, these PPF concentrations induced heart arrhythmia, although no changes in heart rate were observed. Embryos exposed to 0.01 mg/L showed reduced body and heart mass, crown-rump length, and thoracic perimeter, while head circumference was reduced in both exposed groups. Together, combining morphological, molecular, and physiological parameters, this study showed the cardiotoxic effects of PPF exposure and elucidated its impacts on embryonic growth.
{"title":"Exposure to Pyriproxyfen Impacts Heart Development Causing Tissue and Cellular Impairments, Heart Arrhythmia and Reduced Embryonic Growth.","authors":"Maria Fernanda Conte Bernhardt, Nathália Ronconi-Krüger, Evelise Maria Nazari","doi":"10.1007/s12012-024-09944-4","DOIUrl":"10.1007/s12012-024-09944-4","url":null,"abstract":"<p><p>In recent years, concerns have been raised regarding the safety of exposure to pyriproxyfen (PPF), a larvicide commonly used in drinking water reservoirs to control populations of disease-vector mosquitoes for human safety. These concerns are focused mainly on exposure by pregnant women, since studies have shown deleterious effects of PPF on embryonic development, mainly addressing the central nervous system. However, since previous studies showed reduced growth in embryos exposed to PPF, we hypothesize that PPF exposure impairs the cardiovascular system, responsible for ensuring appropriate blood supply, which leads to stunted growth. This study aimed to investigate the impact of PPF exposure on heart ventricular morphology, its influence on cell proliferation and apoptosis, as well as assess the impact on the functionality of the heart and on embryonic growth. Chicken embryos were used as a model and two sublethal concentrations were tested: 0.01 mg/L and 10 mg/L PPF. Thinning of cardiac tissue was evident in heart structures at 10 mg/L PPF. Furthermore, DNA double-strand breaks and reduced cell proliferation were observed, combined with decreased apoptosis suggesting cell cycle arrest, especially in the left ventricle for both concentrations. In addition, these PPF concentrations induced heart arrhythmia, although no changes in heart rate were observed. Embryos exposed to 0.01 mg/L showed reduced body and heart mass, crown-rump length, and thoracic perimeter, while head circumference was reduced in both exposed groups. Together, combining morphological, molecular, and physiological parameters, this study showed the cardiotoxic effects of PPF exposure and elucidated its impacts on embryonic growth.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"85-96"},"PeriodicalIF":3.4,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615330","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}
Doxorubicin (DOX) has a limitation in clinical oncology due to its dose-dependent cardiotoxicity. Thiolutin (THL) can undermine DOX-induced cardiomyocyte injury by inhibiting the NLRP3 inflammasome activation, yet the efficacy of THL in DOX-induced cardiotoxicity (DOXIC) needs to be validated in animal models. DOX-induced mice were treated with THL to evaluate the efficacy of THL. Relative NLRP3 mRNA levels were determined by quantitative PCR. Blood samples were collected from diffuse large B-cell lymphoma (DLBCL) patients with or without DOXIC to validate serum levels of cTnT, IL-1β, CRP, BNP, and IL-18 by enzyme-linked immunosorbent assay. Apoptosis and pyroptosis-related protein levels were analyzed by western blot. Cardiac function and histopathological changes were determined by echocardiography, HE, Masson's, and wheat germ agglutinin staining. In clinical samples, NLRP3 mRNA and/or protein levels were also markedly heightened in peripheral blood mononuclear cells and serum samples from DOXIC patients, along with higher concentrations of IL-18, cTnT, and IL-1β. Importantly, cTnT possessed a positive correlation with NLRP3 mRNA, IL-1β, and IL-18. Moreover, cTnT possessed a positive correlation with NLRP3 mRNA, IL-1β, and IL-18 levels, suggesting a potential link between DOXIC and NLRP3 inflammasome. The outcomes demonstrated that THL reduced LVEF and LVFS, as well as elevated LVESD and LVEDD in DOX-challenged mice, accompanied by elevated serum concentrations of cTnT, CRP, and BNP. In addition, THL attenuated DOX-induced myocardial hypertrophy and cardiac fibrosis in mice, in conjunction with attenuation of DOX-induced upregulation of C-caspase3, Bax, NLRP3, C-caspase-1/Pro-caspase, GSDMD-N/GSDMD, IL-1β, and IL-18 in heart or serum samples. In conclusion, our data supported that THL alleviates the cardiotoxic effects of DOX and suppresses NLRP3 inflammasome in the mouse model, suggesting that THL as a potential drug for DOXIC.
阿霉素(DOX)由于其剂量依赖性的心脏毒性,在临床肿瘤学中具有局限性。硫柳素(THL)可以通过抑制NLRP3炎性体的激活来破坏dox诱导的心肌细胞损伤,但THL在dox诱导的心脏毒性(DOXIC)中的作用需要在动物模型中得到验证。用四氢大麻酚治疗dox诱导小鼠,观察其疗效。定量PCR检测NLRP3 mRNA的相对表达水平。收集弥漫性大b细胞淋巴瘤(DLBCL)患者的血液样本,通过酶联免疫吸附试验验证血清cTnT、IL-1β、CRP、BNP和IL-18的水平。western blot检测细胞凋亡及焦热相关蛋白水平。通过超声心动图、HE、Masson染色和小麦胚芽凝集素染色测定心功能和组织病理学变化。在临床样本中,DOXIC患者外周血单个核细胞和血清样本中NLRP3 mRNA和/或蛋白水平也显著升高,同时IL-18、cTnT和IL-1β浓度升高。重要的是,cTnT与NLRP3 mRNA、IL-1β和IL-18呈正相关。此外,cTnT与NLRP3 mRNA、IL-1β和IL-18水平呈正相关,表明DOXIC与NLRP3炎性体之间存在潜在联系。结果表明,在dox刺激小鼠中,THL可降低LVEF和LVFS,升高LVESD和LVEDD,并伴有血清cTnT、CRP和BNP浓度升高。此外,THL还能减弱dox诱导的小鼠心肌肥大和心脏纤维化,同时还能减弱dox诱导的心脏或血清样品中C-caspase3、Bax、NLRP3、C-caspase3 -1/Pro-caspase、GSDMD- n /GSDMD、IL-1β和IL-18的上调。综上所述,我们的数据支持THL在小鼠模型中减轻DOX的心脏毒性作用并抑制NLRP3炎性体,提示THL是治疗DOXIC的潜在药物。
{"title":"Thiolutin Alleviates Cardiotoxic Effects of Doxorubicin by Suppressing NLRP3 Inflammasome in the Mouse Model.","authors":"Wenyuan Cai, Tingting Teng, Xiaoyan Wang, Baihong Li, Xin Gu, Yafeng Zhou","doi":"10.1007/s12012-024-09947-1","DOIUrl":"https://doi.org/10.1007/s12012-024-09947-1","url":null,"abstract":"<p><p>Doxorubicin (DOX) has a limitation in clinical oncology due to its dose-dependent cardiotoxicity. Thiolutin (THL) can undermine DOX-induced cardiomyocyte injury by inhibiting the NLRP3 inflammasome activation, yet the efficacy of THL in DOX-induced cardiotoxicity (DOXIC) needs to be validated in animal models. DOX-induced mice were treated with THL to evaluate the efficacy of THL. Relative NLRP3 mRNA levels were determined by quantitative PCR. Blood samples were collected from diffuse large B-cell lymphoma (DLBCL) patients with or without DOXIC to validate serum levels of cTnT, IL-1β, CRP, BNP, and IL-18 by enzyme-linked immunosorbent assay. Apoptosis and pyroptosis-related protein levels were analyzed by western blot. Cardiac function and histopathological changes were determined by echocardiography, HE, Masson's, and wheat germ agglutinin staining. In clinical samples, NLRP3 mRNA and/or protein levels were also markedly heightened in peripheral blood mononuclear cells and serum samples from DOXIC patients, along with higher concentrations of IL-18, cTnT, and IL-1β. Importantly, cTnT possessed a positive correlation with NLRP3 mRNA, IL-1β, and IL-18. Moreover, cTnT possessed a positive correlation with NLRP3 mRNA, IL-1β, and IL-18 levels, suggesting a potential link between DOXIC and NLRP3 inflammasome. The outcomes demonstrated that THL reduced LVEF and LVFS, as well as elevated LVESD and LVEDD in DOX-challenged mice, accompanied by elevated serum concentrations of cTnT, CRP, and BNP. In addition, THL attenuated DOX-induced myocardial hypertrophy and cardiac fibrosis in mice, in conjunction with attenuation of DOX-induced upregulation of C-caspase3, Bax, NLRP3, C-caspase-1/Pro-caspase, GSDMD-N/GSDMD, IL-1β, and IL-18 in heart or serum samples. In conclusion, our data supported that THL alleviates the cardiotoxic effects of DOX and suppresses NLRP3 inflammasome in the mouse model, suggesting that THL as a potential drug for DOXIC.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2024-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142812045","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 : 2024-12-01Epub Date: 2024-10-14DOI: 10.1007/s12012-024-09931-9
Yue Ge, Maliha S Nash, Witold M Winnik, Maribel Bruno, William T Padgett, Rachel D Grindstaff, Mehdi S Hazari, Aimen K Farraj
Inhalation of ambient particulate matter (PM) and ozone (O3) has been associated with increased cardiovascular morbidity and mortality. However, the interactive effects of PM and O3 on cardiac dysfunction and disease have not been thoroughly examined, especially at a proteomic level. The purpose of this study was to identify and compare proteome changes in spontaneously hypertensive (SH) rats co-exposed to concentrated ambient particulates (CAPs) and O3, with a focus on investigating inflammatory and metabolic pathways, which are the two major ones implicated in the pathophysiology of cardiac dysfunction. For this, we measured and compared changes in expression status of 9 critical pro- and anti-inflammatory cytokines using multiplexed ELISA and 450 metabolic proteins involved in ATP production, oxidative phosphorylation, cytoskeletal organization, and stress response using two-dimensional electrophoresis (2-DE) and mass spectrometry (MS) in cardiac tissue of SH rats exposed to CAPs alone, O3 alone, and CAPs + O3. Proteomic expression profiling revealed that CAPs alone, O3 alone, and CAPs + O3 differentially altered protein expression patterns, and utilized divergent mechanisms to affect inflammatory and metabolic pathways and responses. Ingenuity Pathway Analysis (IPA) of the proteomic data demonstrated that the metabolic protein network centered by gap junction alpha-1 protein (GJA 1) was interconnected with the inflammatory cytokine network centered by nuclear factor kappa beta (NF-kB) potentially suggesting inflammation-induced alterations in metabolic pathways, or vice versa, collectively contributing to the development of cardiac dysfunction in response to CAPs and O3 exposure. These findings may enhance understanding of the pathophysiology of cardiac dysfunction induced by air pollution and provide testable hypotheses regarding mechanisms of action.
{"title":"Proteomics Reveals Divergent Cardiac Inflammatory and Metabolic Responses After Inhalation of Ambient Particulate Matter With or Without Ozone.","authors":"Yue Ge, Maliha S Nash, Witold M Winnik, Maribel Bruno, William T Padgett, Rachel D Grindstaff, Mehdi S Hazari, Aimen K Farraj","doi":"10.1007/s12012-024-09931-9","DOIUrl":"10.1007/s12012-024-09931-9","url":null,"abstract":"<p><p>Inhalation of ambient particulate matter (PM) and ozone (O<sub>3</sub>) has been associated with increased cardiovascular morbidity and mortality. However, the interactive effects of PM and O<sub>3</sub> on cardiac dysfunction and disease have not been thoroughly examined, especially at a proteomic level. The purpose of this study was to identify and compare proteome changes in spontaneously hypertensive (SH) rats co-exposed to concentrated ambient particulates (CAPs) and O<sub>3</sub>, with a focus on investigating inflammatory and metabolic pathways, which are the two major ones implicated in the pathophysiology of cardiac dysfunction. For this, we measured and compared changes in expression status of 9 critical pro- and anti-inflammatory cytokines using multiplexed ELISA and 450 metabolic proteins involved in ATP production, oxidative phosphorylation, cytoskeletal organization, and stress response using two-dimensional electrophoresis (2-DE) and mass spectrometry (MS) in cardiac tissue of SH rats exposed to CAPs alone, O<sub>3</sub> alone, and CAPs + O<sub>3</sub>. Proteomic expression profiling revealed that CAPs alone, O<sub>3</sub> alone, and CAPs + O<sub>3</sub> differentially altered protein expression patterns, and utilized divergent mechanisms to affect inflammatory and metabolic pathways and responses. Ingenuity Pathway Analysis (IPA) of the proteomic data demonstrated that the metabolic protein network centered by gap junction alpha-1 protein (GJA 1) was interconnected with the inflammatory cytokine network centered by nuclear factor kappa beta (NF-kB) potentially suggesting inflammation-induced alterations in metabolic pathways, or vice versa, collectively contributing to the development of cardiac dysfunction in response to CAPs and O<sub>3</sub> exposure. These findings may enhance understanding of the pathophysiology of cardiac dysfunction induced by air pollution and provide testable hypotheses regarding mechanisms of action.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1348-1363"},"PeriodicalIF":4.3,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142458727","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 : 2024-12-01Epub Date: 2024-10-04DOI: 10.1007/s12012-024-09926-6
Hao Wu, Yan-Nan Che, Qi Lan, Yi-Xiang He, Ping Liu, Ming-Tai Chen, Li Dong, Meng-Nan Liu
The Hippo-yes-associated protein (YAP) signaling pathway plays a crucial role in cell proliferation, differentiation, and death. It is known to have impact on the progression and development of cardiovascular diseases (CVDs) as well as in the regeneration of cardiomyocytes (CMs). However, further research is needed to understand the molecular mechanisms by which the Hippo-YAP pathway affects the pathological processes of CVDs in order to evaluate its potential clinical applications. In this review, we have summarized the recent findings on the role of the Hippo-YAP pathway in CVDs such as myocardial infarction, heart failure, and cardiomyopathy, as well as its in CM development. This review calls attention to the potential roles of the Hippo-YAP pathway as a relevant target for the future treatment of CVDs.
{"title":"The Multifaceted Roles of Hippo-YAP in Cardiovascular Diseases.","authors":"Hao Wu, Yan-Nan Che, Qi Lan, Yi-Xiang He, Ping Liu, Ming-Tai Chen, Li Dong, Meng-Nan Liu","doi":"10.1007/s12012-024-09926-6","DOIUrl":"10.1007/s12012-024-09926-6","url":null,"abstract":"<p><p>The Hippo-yes-associated protein (YAP) signaling pathway plays a crucial role in cell proliferation, differentiation, and death. It is known to have impact on the progression and development of cardiovascular diseases (CVDs) as well as in the regeneration of cardiomyocytes (CMs). However, further research is needed to understand the molecular mechanisms by which the Hippo-YAP pathway affects the pathological processes of CVDs in order to evaluate its potential clinical applications. In this review, we have summarized the recent findings on the role of the Hippo-YAP pathway in CVDs such as myocardial infarction, heart failure, and cardiomyopathy, as well as its in CM development. This review calls attention to the potential roles of the Hippo-YAP pathway as a relevant target for the future treatment of CVDs.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1410-1427"},"PeriodicalIF":3.4,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142371053","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 : 2024-12-01Epub Date: 2024-09-22DOI: 10.1007/s12012-024-09921-x
Mohammad Mahdi Dabbaghi, Hesan Soleimani Roudi, Rozhan Safaei, Vafa Baradaran Rahimi, Mohammad Reza Fadaei, Vahid Reza Askari
Tanshinone, a natural compound found in the roots of Salvia miltiorrhiza, has been shown to possess various pharmacological properties, including anti-inflammatory, antioxidant, and cardiovascular protective effects. This article aims to review the literature on the cardiovascular protective effects of tanshinone and its underlying mechanisms. Tanshinone has been demonstrated to improve cardiac function, reduce oxidative stress, and inhibit inflammation in various animal models of cardiovascular diseases. Additionally, it has been shown to regulate multiple signaling pathways involved in the pathogenesis of cardiovascular diseases, such as the PI3K/AKT, MAPK, and NF-κB pathways. Clinical studies have also suggested that tanshinone may have therapeutic potential for treating cardiovascular diseases. In conclusion, tanshinone has emerged as a promising natural compound with significant cardiovascular protective effects, and further research is warranted to explore its potential clinical applications.
{"title":"Unveiling the Mechanism of Protective Effects of Tanshinone as a New Fighter Against Cardiovascular Diseases: A Systematic Review.","authors":"Mohammad Mahdi Dabbaghi, Hesan Soleimani Roudi, Rozhan Safaei, Vafa Baradaran Rahimi, Mohammad Reza Fadaei, Vahid Reza Askari","doi":"10.1007/s12012-024-09921-x","DOIUrl":"10.1007/s12012-024-09921-x","url":null,"abstract":"<p><p>Tanshinone, a natural compound found in the roots of Salvia miltiorrhiza, has been shown to possess various pharmacological properties, including anti-inflammatory, antioxidant, and cardiovascular protective effects. This article aims to review the literature on the cardiovascular protective effects of tanshinone and its underlying mechanisms. Tanshinone has been demonstrated to improve cardiac function, reduce oxidative stress, and inhibit inflammation in various animal models of cardiovascular diseases. Additionally, it has been shown to regulate multiple signaling pathways involved in the pathogenesis of cardiovascular diseases, such as the PI<sub>3</sub>K/AKT, MAPK, and NF-κB pathways. Clinical studies have also suggested that tanshinone may have therapeutic potential for treating cardiovascular diseases. In conclusion, tanshinone has emerged as a promising natural compound with significant cardiovascular protective effects, and further research is warranted to explore its potential clinical applications.</p>","PeriodicalId":9570,"journal":{"name":"Cardiovascular Toxicology","volume":" ","pages":"1467-1509"},"PeriodicalIF":4.3,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280634","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}