首页 > 最新文献

Antioxidants & redox signaling最新文献

英文 中文
Redox Regulation of Microvascular Physiology and Pathophysiology: Insights into Therapeutic Strategies and Limitations. 微血管生理学和病理生理学的氧化还原调节:对治疗策略和局限性的见解。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-01 Epub Date: 2025-09-29 DOI: 10.1177/15230864251372607
David A Bulger, Zhan Zhang, Ruinan Hu, Esha K Dave, Puja K Mehta, Kathy K Griendling, Alejandra Valdivia

Significance: Oxidative mechanisms contribute to both vascular function and pathogenesis of many diseases, but their role in the microvasculature remains poorly understood. Recent Advances: The role of reactive oxygen and reactive nitrogen species (ROS/RNS) in the vasculature has been well-established for years. Our knowledge of microvascular responses to ROS/RNS has relied on extrapolation of studies performed in large vessels or cultured endothelial cells from large vessels. In healthy tissue, ROS/RNS are implicated in microvascular cell survival and death, angiogenesis, vasodilation, and barrier function, and, in disease, they contribute to increased permeability, leukocyte extravasation, and inflammation. Redox-mediated microvascular dysfunction underlies a multitude of conditions, including cardiovascular diseases, autoimmune diseases, infectious diseases, hemoglobinopathies, inflammatory diseases, vasculitides, and metabolic diseases. Critical Issues: New single-cell RNA sequencing studies reveal that endothelial cells from different vascular beds have unique gene signatures. Moreover, microvessels respond differently than large vessels, yet findings are frequently extrapolated across vascular beds. Technical challenges have limited our ability to reliably link alterations in ROS/RNS levels to microvascular outcomes. Moreover, successful therapeutics targeting redox signaling in general and in the microvasculature in particular are lacking. While numerous associations exist between common diseases and the microvasculature, the precise contribution of redox-mediated microvascular dysfunction to disease pathogenesis has been challenging. Future Directions: Additional research in organ-specific microvasculature focusing on the redox mechanisms underlying microvascular function and dysfunction is needed, as well as the development of new targeted therapeutics that can be locally delivered. Comparison of redox responses between different diseases may uncover general mechanisms to exploit therapeutically. Antioxid. Redox Signal. 43, 566-621.

意义:氧化机制参与许多疾病的血管功能和发病机制,但其在微血管系统中的作用尚不清楚。近年来,活性氧和活性氮(ROS/RNS)在血管系统中的作用已经得到了广泛的研究。我们对微血管对ROS/RNS反应的了解依赖于在大血管或培养的大血管内皮细胞中进行的研究的推断。在健康组织中,ROS/RNS与微血管细胞的存活和死亡、血管生成、血管舒张和屏障功能有关,在疾病中,它们有助于通透性增加、白细胞外渗和炎症。氧化还原介导的微血管功能障碍是多种疾病的基础,包括心血管疾病、自身免疫性疾病、传染病、血红蛋白病、炎症性疾病、血管炎和代谢性疾病。关键问题:新的单细胞RNA测序研究表明,来自不同血管床的内皮细胞具有独特的基因特征。此外,微血管的反应与大血管不同,但研究结果经常被推断为跨血管床。技术上的挑战限制了我们将ROS/RNS水平的改变与微血管结果可靠地联系起来的能力。此外,目前还缺乏针对氧化还原信号的成功治疗方法,特别是针对微血管的治疗方法。虽然常见疾病与微血管系统之间存在许多关联,但氧化还原介导的微血管功能障碍对疾病发病机制的确切贡献一直具有挑战性。未来方向:需要对微血管功能和功能障碍的氧化还原机制进行更多的器官特异性微血管研究,以及开发新的局部靶向治疗方法。比较不同疾病之间的氧化还原反应可能揭示一般机制,以开发治疗。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Redox Regulation of Microvascular Physiology and Pathophysiology: Insights into Therapeutic Strategies and Limitations.","authors":"David A Bulger, Zhan Zhang, Ruinan Hu, Esha K Dave, Puja K Mehta, Kathy K Griendling, Alejandra Valdivia","doi":"10.1177/15230864251372607","DOIUrl":"10.1177/15230864251372607","url":null,"abstract":"<p><p><b><i>Significance:</i></b> Oxidative mechanisms contribute to both vascular function and pathogenesis of many diseases, but their role in the microvasculature remains poorly understood. <b><i>Recent Advances:</i></b> The role of reactive oxygen and reactive nitrogen species (ROS/RNS) in the vasculature has been well-established for years. Our knowledge of microvascular responses to ROS/RNS has relied on extrapolation of studies performed in large vessels or cultured endothelial cells from large vessels. In healthy tissue, ROS/RNS are implicated in microvascular cell survival and death, angiogenesis, vasodilation, and barrier function, and, in disease, they contribute to increased permeability, leukocyte extravasation, and inflammation. Redox-mediated microvascular dysfunction underlies a multitude of conditions, including cardiovascular diseases, autoimmune diseases, infectious diseases, hemoglobinopathies, inflammatory diseases, vasculitides, and metabolic diseases. <b><i>Critical Issues:</i></b> New single-cell RNA sequencing studies reveal that endothelial cells from different vascular beds have unique gene signatures. Moreover, microvessels respond differently than large vessels, yet findings are frequently extrapolated across vascular beds. Technical challenges have limited our ability to reliably link alterations in ROS/RNS levels to microvascular outcomes. Moreover, successful therapeutics targeting redox signaling in general and in the microvasculature in particular are lacking. While numerous associations exist between common diseases and the microvasculature, the precise contribution of redox-mediated microvascular dysfunction to disease pathogenesis has been challenging. <b><i>Future Directions:</i></b> Additional research in organ-specific microvasculature focusing on the redox mechanisms underlying microvascular function and dysfunction is needed, as well as the development of new targeted therapeutics that can be locally delivered. Comparison of redox responses between different diseases may uncover general mechanisms to exploit therapeutically. <i>Antioxid. Redox Signal.</i> 43, 566-621.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"566-621"},"PeriodicalIF":6.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145184583","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of Hydrogen Sulfide Regulation of Programmed Cell Death: Implications for Cardiovascular Diseases. 硫化氢调控程序性细胞死亡的作用:对心血管疾病的影响。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-01 DOI: 10.1177/15230864251386449
Qing-Bo Lu, Xue-Xue Zhu, Guo Chen, Jia-Bao Su, Chen-Yang Zhao, An-Jing Xu, Jin-Song Bian, Hai-Jian Sun

Significance: Hydrogen sulfide (H2S) is an important signaling molecule involved in cardiovascular diseases (CVDs). Although it is important, the precise mechanisms underlying the diverse functions of H2S in CVDs are not known and need to be elucidated. Recent Advances: Studies have shown the importance of different programmed cell death (PCD) modalities, such as NETosis, apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, in the pathogenesis of CVDs. An overview of the role of H2S in regulating PCD in diabetic cardiomyopathy (DCM), cardiac hypertrophy and fibrosis, hypertension, heart failure, atherosclerosis and myocardial ischemia/reperfusion injury, might provide a better understanding of the cardiovascular effects of H2S. Critical Issues: The mechanisms by which H2S modulates each type of PCD in CVD patients need to be elucidated. The differences in the effects of H2S on PCD modalities in different cardiovascular cell types, such as cardiomyocytes, endothelial cells, smooth muscle cells, and immune cells, require further evidence. Future Directions: Future studies should focus on the mechanism by which H2S affects distinct PCD pathways. Whether H2S acts as a switch between different PCD pathways under stress or disease conditions needs to be determined. H2S might regulate the temporal and spatial overlapping PCD pathways in CVDs. Single-cell RNA sequences, spatial transcriptomics, and live-cell imaging are needed to map PCD events regulated by H2S. Innovation: In this review, we summarized the regulatory effects of H2S on signaling pathways related to PCD in patients with CVDs. Understanding these mechanisms is crucial for elucidating the pathophysiological roles of H2S in CVDs. Antioxid. Redox Signal. 43, 637-690.

意义:硫化氢(H2S)是参与心血管疾病(cvd)的重要信号分子。虽然它很重要,但H2S在cvd中多种功能的确切机制尚不清楚,需要阐明。最新进展:研究表明,不同的程序性细胞死亡(PCD)模式,如NETosis、凋亡、necroptosis、pyroptosis、ferroptosis和cuprotosis,在cvd的发病机制中的重要性。概述H2S在糖尿病性心肌病(DCM)、心脏肥大和纤维化、高血压、心力衰竭、动脉粥样硬化和心肌缺血/再灌注损伤中调控PCD的作用,可能有助于更好地理解H2S对心血管的影响。关键问题:需要阐明H2S调节CVD患者每种类型PCD的机制。H2S对不同心血管细胞类型(如心肌细胞、内皮细胞、平滑肌细胞和免疫细胞)PCD模式的影响差异,需要进一步的证据。未来研究方向:未来的研究应关注H2S影响不同PCD途径的机制。在压力或疾病条件下,H2S是否作为不同PCD途径之间的开关需要确定。H2S可能调节cvd中PCD通路的时空重叠。需要单细胞RNA序列、空间转录组学和活细胞成像来绘制H2S调控的PCD事件。创新:在这篇综述中,我们总结了H2S对心血管疾病患者PCD相关信号通路的调节作用。了解这些机制对于阐明H2S在心血管疾病中的病理生理作用至关重要。Antioxid。氧化还原信号,43,637-690。
{"title":"Role of Hydrogen Sulfide Regulation of Programmed Cell Death: Implications for Cardiovascular Diseases.","authors":"Qing-Bo Lu, Xue-Xue Zhu, Guo Chen, Jia-Bao Su, Chen-Yang Zhao, An-Jing Xu, Jin-Song Bian, Hai-Jian Sun","doi":"10.1177/15230864251386449","DOIUrl":"https://doi.org/10.1177/15230864251386449","url":null,"abstract":"<p><p><b><i>Significance:</i></b> Hydrogen sulfide (H<sub>2</sub>S) is an important signaling molecule involved in cardiovascular diseases (CVDs). Although it is important, the precise mechanisms underlying the diverse functions of H<sub>2</sub>S in CVDs are not known and need to be elucidated. <b><i>Recent Advances:</i></b> Studies have shown the importance of different programmed cell death (PCD) modalities, such as NETosis, apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, in the pathogenesis of CVDs. An overview of the role of H<sub>2</sub>S in regulating PCD in diabetic cardiomyopathy (DCM), cardiac hypertrophy and fibrosis, hypertension, heart failure, atherosclerosis and myocardial ischemia/reperfusion injury, might provide a better understanding of the cardiovascular effects of H<sub>2</sub>S. <b><i>Critical Issues:</i></b> The mechanisms by which H<sub>2</sub>S modulates each type of PCD in CVD patients need to be elucidated. The differences in the effects of H<sub>2</sub>S on PCD modalities in different cardiovascular cell types, such as cardiomyocytes, endothelial cells, smooth muscle cells, and immune cells, require further evidence. <b><i>Future Directions:</i></b> Future studies should focus on the mechanism by which H<sub>2</sub>S affects distinct PCD pathways. Whether H<sub>2</sub>S acts as a switch between different PCD pathways under stress or disease conditions needs to be determined. H<sub>2</sub>S might regulate the temporal and spatial overlapping PCD pathways in CVDs. Single-cell RNA sequences, spatial transcriptomics, and live-cell imaging are needed to map PCD events regulated by H<sub>2</sub>S. <b><i>Innovation:</i></b> In this review, we summarized the regulatory effects of H<sub>2</sub>S on signaling pathways related to PCD in patients with CVDs. Understanding these mechanisms is crucial for elucidating the pathophysiological roles of H<sub>2</sub>S in CVDs. <i>Antioxid. Redox Signal.</i> 43, 637-690.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":"43 10-12","pages":"637-690"},"PeriodicalIF":6.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145367452","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reprogramming Iron Metabolism via the RIG-I/c-Myc/FTH Axis Mitigates Renal Ischemia-Reperfusion Injury. 通过rig - 1 /c-Myc/FTH轴重编程铁代谢减轻肾缺血再灌注损伤
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-01 Epub Date: 2025-08-22 DOI: 10.1177/15230864251369883
Yulu Zhang, Jia Xing, Li Yao, Yu Zou, Hui Peng, Xiling Yi, Lifang Bai, Yang Yu, Hanzhe Liu, Xue Li, Xiaoyue Zhai

Aims: Iron metabolism disorders are critical in the pathogenesis of acute kidney ischemia-reperfusion injury (IRI). However, the molecular mechanisms driving these disturbances remain poorly understood. Results: In IRI mouse kidneys, pathological alterations, iron metabolism disruptions, and functional impairments were observed. Retinoic acid-inducible gene-I (RIG-I), transcription factor c-Myc, and ferritin heavy chain (FTH) exhibited elevated expression and colocalization in tubular epithelial cells, accompanied by decreased glutathione peroxidase 4 (GPX4) level and evidence of ferroptosis. Further in vitro studies revealed that RIG-I promoted c-Myc activation. The latter demonstrated its positive regulation of FTH transcription by chromatin immunoprecipitation assays and c-Myc siRNA experiments. Interestingly, FTH overexpression resulted in elevated levels of RIG-I, transferrin receptor, ferroportin, and nuclear receptor coactivator 4. Ultimately, the c-Myc inhibitor 10058-F4 reversed all adverse alterations and demonstrated a protective role in IRI mouse kidneys and mouse kidney tubule cells subjected to the ferroptosis inducer erastin, RIG-I agonist, or hypoxia/reoxygenation. This reversal was reflected in improved renal morphology and function, balanced iron metabolism, increased GPX4 level, decreased 4-hydroxynonenal level, reduced inflammatory cell infiltration, interleukin-1 beta release, and kidney injury molecule 1 expression. Innovation: This study proposes a novel mechanism in which c-Myc is activated by elevated RIG-I in IRI kidneys and positively regulates FTH transcription, therefore involving iron metabolism disorders. Conclusions: The RIG-I, c-Myc, and FTH disrupt iron homeostasis, and the c-Myc inhibition stabilizes iron metabolism and mitigates oxidative stress, suggesting a potential therapeutic target in IRI. Antioxid. Redox Signal. 43, 622-636. [Figure: see text].

目的:铁代谢紊乱在急性肾缺血再灌注损伤(IRI)的发病机制中起关键作用。然而,驱动这些干扰的分子机制仍然知之甚少。结果:在IRI小鼠肾脏中,观察到病理改变、铁代谢中断和功能损伤。维甲酸诱导基因i (RIG-I)、转录因子c-Myc和铁蛋白重链(FTH)在小管上皮细胞中的表达和共定位升高,并伴有谷胱甘肽过氧化物酶4 (GPX4)水平下降和铁下垂的证据。进一步的体外研究表明,RIG-I促进了c-Myc的活化。后者通过染色质免疫沉淀试验和c-Myc siRNA实验证明其对FTH转录有正向调节作用。有趣的是,FTH过表达导致rig - 1、转铁蛋白受体、铁转运蛋白和核受体共激活因子4水平升高。最终,c-Myc抑制剂10058-F4逆转了所有不利的改变,并证明了IRI小鼠肾脏和小鼠肾小管细胞受到铁凋亡诱导剂erastin、RIG-I激动剂或缺氧/再氧化的保护作用。这种逆转表现为肾脏形态和功能改善,铁代谢平衡,GPX4水平升高,4-羟基烯醛水平降低,炎症细胞浸润、白细胞介素-1 β释放和肾损伤分子1表达减少。创新:本研究提出了一种新的机制,在IRI肾脏中,c-Myc被RIG-I升高激活,并积极调节FTH转录,因此涉及铁代谢紊乱。结论:RIG-I、c-Myc和FTH破坏铁稳态,抑制c-Myc稳定铁代谢并减轻氧化应激,提示IRI的潜在治疗靶点。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Reprogramming Iron Metabolism via the RIG-I/c-Myc/FTH Axis Mitigates Renal Ischemia-Reperfusion Injury.","authors":"Yulu Zhang, Jia Xing, Li Yao, Yu Zou, Hui Peng, Xiling Yi, Lifang Bai, Yang Yu, Hanzhe Liu, Xue Li, Xiaoyue Zhai","doi":"10.1177/15230864251369883","DOIUrl":"10.1177/15230864251369883","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Iron metabolism disorders are critical in the pathogenesis of acute kidney ischemia-reperfusion injury (IRI). However, the molecular mechanisms driving these disturbances remain poorly understood. <b><i>Results:</i></b> In IRI mouse kidneys, pathological alterations, iron metabolism disruptions, and functional impairments were observed. Retinoic acid-inducible gene-I (RIG-I), transcription factor c-Myc, and ferritin heavy chain (FTH) exhibited elevated expression and colocalization in tubular epithelial cells, accompanied by decreased glutathione peroxidase 4 (GPX4) level and evidence of ferroptosis. Further <i>in vitro</i> studies revealed that RIG-I promoted c-Myc activation. The latter demonstrated its positive regulation of FTH transcription by chromatin immunoprecipitation assays and c-Myc siRNA experiments. Interestingly, FTH overexpression resulted in elevated levels of RIG-I, transferrin receptor, ferroportin, and nuclear receptor coactivator 4. Ultimately, the c-Myc inhibitor 10058-F4 reversed all adverse alterations and demonstrated a protective role in IRI mouse kidneys and mouse kidney tubule cells subjected to the ferroptosis inducer erastin, RIG-I agonist, or hypoxia/reoxygenation. This reversal was reflected in improved renal morphology and function, balanced iron metabolism, increased GPX4 level, decreased 4-hydroxynonenal level, reduced inflammatory cell infiltration, interleukin-1 beta release, and kidney injury molecule 1 expression. <b><i>Innovation:</i></b> This study proposes a novel mechanism in which c-Myc is activated by elevated RIG-I in IRI kidneys and positively regulates FTH transcription, therefore involving iron metabolism disorders. <b><i>Conclusions:</i></b> The RIG-I, c-Myc, and FTH disrupt iron homeostasis, and the c-Myc inhibition stabilizes iron metabolism and mitigates oxidative stress, suggesting a potential therapeutic target in IRI. <i>Antioxid. Redox Signal.</i> 43, 622-636. [Figure: see text].</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"622-636"},"PeriodicalIF":6.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144939846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
High Incidence of Lethal Ventricular Arrhythmia-Sudden Cardiac Death in Early Myocardial Ischemia: Critical Roles of Cross-Regulation Between Stresses and Calcium Imbalance. 心肌缺血早期致死性室性心律失常-心源性猝死的高发:应激与钙失衡交叉调节的关键作用
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-01 Epub Date: 2025-09-15 DOI: 10.1177/15230864251372589
Xiaojuan Zhang, Mengxuan Zhang, Ye Zhang, Wei Zhang, Huishan Liang, Junyao Lv, Xudong Xiao, Guanghui Zhu, Xiaojun Yu, Minchao Lai, Dian Wang

Aims: Early myocardial ischemia (MI) predisposes to lethal ventricular arrhythmias (LVA) and subsequent sudden cardiac death (SCD). This study aims to elucidate the roles of cross-regulation between oxidative stress, endoplasmic reticulum (ER) stress, and calcium (Ca2+) disturbances in the increased risk of LVA-SCD in early MI. Results: Both clinical and animal model data showed a higher incidence of SCD within 30 min of MI. In MI animals, T-wave alternans and conduction slowing were observed prior to LVA onset. Optical mapping revealed spatiotemporal electrophysiological discordances, including conduction slowing and alternans in both action potentials and Ca2+ transients before LVA, peaking 5-15 min after ischemia onset, with the ischemic zone most affected. Reentrant cycles were observed in isolated MI hearts that developed LVA. SCD animals exhibited elevated mitochondrial and cytosolic reactive oxygen species and Ca2+, mitochondrial damage, ER stressors upregulation, and activation of the Ca2+/calmodulin-dependent protein kinases (oxidized)-RyR2, ryanodine receptor 2 (CaMKII-RyR2) pathway. These results were partly validated in hypoxic and undernourished myocytes. Targeted interventions, such as MitoTEMPO to mitigate oxidative stress, 4-phenyl butyric acid to inhibit ER stress, and dantrolene or RyR2-S2814A to suppress Ca2+ leakage, attenuated disturbances and reduced SCD incidence. Innovation and Conclusion: We identify a critical 30-min window post-MI, during which redox/ER stress and Ca2 imbalance synergistically drive LVA and SCD via the CaMKII-RyR2 pathway. Targeting this pathway could offer a promising strategy to prevent LVA and SCD in early MI. Antioxid. Redox Signal. 43, 547-565.

目的:早期心肌缺血(MI)易致死性室性心律失常(LVA)和随后的心源性猝死(SCD)。本研究旨在阐明氧化应激、内质网(ER)应激和钙(Ca2+)干扰在心肌梗死早期LVA-SCD风险增加中的交叉调节作用。结果:临床和动物模型数据均显示心肌梗死30分钟内SCD发生率较高。在心肌梗死动物中,在LVA发病前观察到t波交替和传导减慢。光学映射显示时空电生理不一致,包括LVA前动作电位和Ca2+瞬态的传导减慢和交替,在缺血发作后5-15分钟达到峰值,缺血区受影响最大。在发生LVA的离体心肌梗死心脏中观察到再入周期。SCD动物表现出线粒体和细胞质活性氧和Ca2+升高,线粒体损伤,内质网应激因子上调,Ca2+/钙调素依赖性蛋白激酶(氧化)-RyR2, ryanodine受体2 (CaMKII-RyR2)通路激活。这些结果在缺氧和营养不良的肌细胞中得到了部分验证。有针对性的干预措施,如MitoTEMPO减轻氧化应激,4-苯基丁酸抑制内质网应激,丹trolene或RyR2-S2814A抑制Ca2+泄漏,减弱干扰,降低SCD发病率。创新和结论:我们确定了心肌梗死后一个关键的30分钟窗口,在此期间,氧化还原/内质网应激和Ca2失衡通过CaMKII-RyR2途径协同驱动LVA和SCD。靶向这一途径可能为预防早期心肌梗死的LVA和SCD提供一种有希望的策略。氧化还原信号:00000 - 00000。
{"title":"High Incidence of Lethal Ventricular Arrhythmia-Sudden Cardiac Death in Early Myocardial Ischemia: Critical Roles of Cross-Regulation Between Stresses and Calcium Imbalance.","authors":"Xiaojuan Zhang, Mengxuan Zhang, Ye Zhang, Wei Zhang, Huishan Liang, Junyao Lv, Xudong Xiao, Guanghui Zhu, Xiaojun Yu, Minchao Lai, Dian Wang","doi":"10.1177/15230864251372589","DOIUrl":"10.1177/15230864251372589","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Early myocardial ischemia (MI) predisposes to lethal ventricular arrhythmias (LVA) and subsequent sudden cardiac death (SCD). This study aims to elucidate the roles of cross-regulation between oxidative stress, endoplasmic reticulum (ER) stress, and calcium (Ca<sup>2+</sup>) disturbances in the increased risk of LVA-SCD in early MI. <b><i>Results:</i></b> Both clinical and animal model data showed a higher incidence of SCD within 30 min of MI. In MI animals, T-wave alternans and conduction slowing were observed prior to LVA onset. Optical mapping revealed spatiotemporal electrophysiological discordances, including conduction slowing and alternans in both action potentials and Ca<sup>2+</sup> transients before LVA, peaking 5-15 min after ischemia onset, with the ischemic zone most affected. Reentrant cycles were observed in isolated MI hearts that developed LVA. SCD animals exhibited elevated mitochondrial and cytosolic reactive oxygen species and Ca<sup>2+</sup>, mitochondrial damage, ER stressors upregulation, and activation of the Ca<sup>2+</sup>/calmodulin-dependent protein kinases (oxidized)-RyR2, ryanodine receptor 2 (CaMKII-RyR2) pathway. These results were partly validated in hypoxic and undernourished myocytes. Targeted interventions, such as MitoTEMPO to mitigate oxidative stress, 4-phenyl butyric acid to inhibit ER stress, and dantrolene or RyR2-S2814A to suppress Ca<sup>2+</sup> leakage, attenuated disturbances and reduced SCD incidence. <b><i>Innovation and Conclusion:</i></b> We identify a critical 30-min window post-MI, during which redox/ER stress and Ca<sup>2</sup> imbalance synergistically drive LVA and SCD <i>via</i> the CaMKII-RyR2 pathway. Targeting this pathway could offer a promising strategy to prevent LVA and SCD in early MI. <i>Antioxid. Redox Signal.</i> 43, 547-565.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"547-565"},"PeriodicalIF":6.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145063353","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oncoviral Infection and the Significance of Reactive Oxygen Species: From Mechanisms to Therapeutic Significance. 肿瘤病毒感染与活性氧的意义:从机制到治疗意义。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-01 Epub Date: 2025-07-08 DOI: 10.1089/ars.2024.0868
Ruixue Sang, Xia Zhao, Ketao Sun, Yan Zhang, Bing Luo

Significance: Reactive oxygen species (ROS) are a double-edged sword in the context of oncoviruses. The effects of ROS on cells depend on the cellular environment, the stage of the disease, and the specific molecular pathways involved. In general, ROS levels in oncovirus-infected cells are usually increased and produce two distinct outcomes on cancer progression and metastasis through multiple mechanisms. Therefore, identifying the relationship between ROS and tumor viruses at the molecular level is essential for cancer prevention and treatment. Recent Advances: ROS play an important role in oncoviral infection and disease progression. The excessive accumulation of ROS induces ferroptosis, which has an important role in tumor therapy and the immune microenvironment, thus providing a theoretical basis for the development of new anticancer treatment strategies. Critical Issues: This review summarizes the complex relationship between ROS and oncoviral infection, with the aim of providing a deeper understanding of tumor pathogenesis and new therapeutic strategies. Future Directions: The relationship between ROS induced by oncoviral infection and host metabolic pathways, including lipids, lipoproteins, amino acids, and polyamines. Understanding how metabolism is reprogrammed in cancer cells may elucidate the impact of these processes on viral infection and tumor progression and help develop effective treatment strategies. Antioxid. Redox Signal. 43, 528-546.

意义:活性氧(ROS)在肿瘤病毒中是一把双刃剑。活性氧对细胞的影响取决于细胞环境、疾病的阶段和所涉及的特定分子途径。通常,在癌病毒感染的细胞中,ROS水平通常升高,并通过多种机制在癌症进展和转移中产生两种不同的结果。因此,在分子水平上明确ROS与肿瘤病毒的关系,对于癌症的预防和治疗至关重要。最新进展:活性氧在肿瘤病毒感染和疾病进展中起重要作用。ROS的过度积累诱导铁下垂,在肿瘤治疗和免疫微环境中具有重要作用,从而为开发新的抗癌治疗策略提供了理论依据。关键问题:本文综述了活性氧与肿瘤病毒感染之间的复杂关系,旨在为深入了解肿瘤发病机制和新的治疗策略提供帮助。未来发展方向:肿瘤病毒感染诱导的活性氧与宿主代谢途径的关系,包括脂质、脂蛋白、氨基酸和多胺。了解癌细胞中的代谢是如何重编程的,可以阐明这些过程对病毒感染和肿瘤进展的影响,并有助于制定有效的治疗策略。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Oncoviral Infection and the Significance of Reactive Oxygen Species: From Mechanisms to Therapeutic Significance.","authors":"Ruixue Sang, Xia Zhao, Ketao Sun, Yan Zhang, Bing Luo","doi":"10.1089/ars.2024.0868","DOIUrl":"10.1089/ars.2024.0868","url":null,"abstract":"<p><p><b><i>Significance:</i></b> Reactive oxygen species (ROS) are a double-edged sword in the context of oncoviruses. The effects of ROS on cells depend on the cellular environment, the stage of the disease, and the specific molecular pathways involved. In general, ROS levels in oncovirus-infected cells are usually increased and produce two distinct outcomes on cancer progression and metastasis through multiple mechanisms. Therefore, identifying the relationship between ROS and tumor viruses at the molecular level is essential for cancer prevention and treatment. <b><i>Recent Advances:</i></b> ROS play an important role in oncoviral infection and disease progression. The excessive accumulation of ROS induces ferroptosis, which has an important role in tumor therapy and the immune microenvironment, thus providing a theoretical basis for the development of new anticancer treatment strategies. <b><i>Critical Issues:</i></b> This review summarizes the complex relationship between ROS and oncoviral infection, with the aim of providing a deeper understanding of tumor pathogenesis and new therapeutic strategies. <b><i>Future Directions:</i></b> The relationship between ROS induced by oncoviral infection and host metabolic pathways, including lipids, lipoproteins, amino acids, and polyamines. Understanding how metabolism is reprogrammed in cancer cells may elucidate the impact of these processes on viral infection and tumor progression and help develop effective treatment strategies. <i>Antioxid. Redox Signal.</i> 43, 528-546.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"528-546"},"PeriodicalIF":6.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144582910","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mitochondrial Ucp4 Ameliorates Motor Disorders by Protecting Cerebellar Purkinje Cells from Oxidative Stress in Intermittent Hypobaric Hypoxia Mice. 线粒体Ucp4通过保护间歇低压缺氧小鼠小脑浦肯野细胞免受氧化应激改善运动障碍
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-01 Epub Date: 2025-07-09 DOI: 10.1089/ars.2024.0853
Fei-Fei Wu, Bo-Zhi Liu, Rui-Qing Wang, Yun-Qiang Huang, Hui Liu, Zi-Wei Ni, Bo-Yang Li, Yu-Ze Sun, Yan-Ling Yang, Ya-Yun Wang

Acute altitude hypoxia is a syndrome that manifests at elevations exceeding 2500 m, posing significant health challenges to individuals who travel or work at high altitudes. Uncoupling proteins are integral proteins located within the mitochondrial inner membrane, playing a crucial role in modulating proton leakage across the mitochondrial membrane. This study investigates the potential role of uncoupling protein 4 (Ucp4) overexpression in an intermittent hypobaric hypoxia (IHH) model and its underlying mechanisms in the cerebellar dyskinesia phenotype. An IHH model was developed using a low-pressure hypoxic chamber, exposing mice to 16 h of hypoxia daily for 5 days. Three mouse strains were used: C57BL/6J, Pcp2Cre; Ucp4fl/fl, and Pcp2Cre; Mito-GFP. Behavioral tests, including rotarod, open field, balance beam, and Morris water maze, were conducted. Ucp4-overexpressing virus was administered to cerebellar lobes 4/5. Mitochondrial morphology was assessed via transmission electron microscopy, 3D reconstruction, and network analysis, while function was evaluated through reactive oxygen species, mitochondrial membrane potential (MMP), glutathione/glutathione disulfide ratio, adenosine triphosphate levels, qPCR, and Western blotting. Results showed that IHH induces hypoactivity without affecting spatial cognition. IHH-induced hypoactivity is linked to Ucp4 upregulation and increased mitochondrial fragmentation in Purkinje cells (PCs), though overall mitochondrial dynamics remain balanced. Ucp4 deficiency exacerbates IHH-induced hypoactivity and mitochondrial fragmentation. Conversely, Ucp4 overexpression in PCs significantly alleviates these effects. Mechanistically, Ucp4 protects PCs by stabilizing MMP and regulating oxidative stress, maintaining mitochondrial integrity. This study reveals that Ucp4 protects cerebellar PCs from oxidative stress in IHH, improving motor function and identifying Ucp4 as a potential therapeutic target for intermittent high-altitude syndrome. Antioxid. Redox Signal. 43, 483-508.

急性高原缺氧是一种在海拔超过2500米时表现出来的综合征,对在高海拔地区旅行或工作的人构成重大的健康挑战。解偶联蛋白是位于线粒体内膜内的完整蛋白,在调节质子穿过线粒体膜的泄漏中起着至关重要的作用。本研究探讨了解偶联蛋白4 (Ucp4)过表达在间歇性低压缺氧(IHH)模型中的潜在作用及其在小脑运动障碍表型中的潜在机制。采用低压缺氧舱建立IHH模型,每天缺氧16小时,持续5天。采用三种小鼠品系:C57BL/6J、Pcp2Cre;Ucp4fl/fl,和pcp2cr;Mito-GFP。行为学测试包括旋转杆、空地、平衡木和Morris水迷宫。过表达ucp4的病毒给药于小脑叶4/5。通过透射电镜、3D重建和网络分析评估线粒体形态,通过活性氧、线粒体膜电位(MMP)、谷胱甘肽/谷胱甘肽二硫比、三磷酸腺苷水平、qPCR和Western blotting评估功能。结果表明,IHH诱导低活动,但不影响空间认知。ihh诱导的低活性与浦肯野细胞(PCs)中Ucp4上调和线粒体断裂增加有关,尽管总体线粒体动力学保持平衡。Ucp4缺乏加剧了ihh诱导的低活性和线粒体断裂。相反,Ucp4在pc中的过表达可显著缓解这些影响。机制上,Ucp4通过稳定MMP和调节氧化应激,维持线粒体完整性来保护pc。本研究揭示了Ucp4保护IHH小脑pc免受氧化应激,改善运动功能,并确定Ucp4是间歇性高原综合征的潜在治疗靶点。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Mitochondrial Ucp4 Ameliorates Motor Disorders by Protecting Cerebellar Purkinje Cells from Oxidative Stress in Intermittent Hypobaric Hypoxia Mice.","authors":"Fei-Fei Wu, Bo-Zhi Liu, Rui-Qing Wang, Yun-Qiang Huang, Hui Liu, Zi-Wei Ni, Bo-Yang Li, Yu-Ze Sun, Yan-Ling Yang, Ya-Yun Wang","doi":"10.1089/ars.2024.0853","DOIUrl":"10.1089/ars.2024.0853","url":null,"abstract":"<p><p>Acute altitude hypoxia is a syndrome that manifests at elevations exceeding 2500 m, posing significant health challenges to individuals who travel or work at high altitudes. Uncoupling proteins are integral proteins located within the mitochondrial inner membrane, playing a crucial role in modulating proton leakage across the mitochondrial membrane. This study investigates the potential role of uncoupling protein 4 (Ucp4) overexpression in an intermittent hypobaric hypoxia (IHH) model and its underlying mechanisms in the cerebellar dyskinesia phenotype. An IHH model was developed using a low-pressure hypoxic chamber, exposing mice to 16 h of hypoxia daily for 5 days. Three mouse strains were used: C57BL/6J, Pcp2<sup>Cre</sup>; Ucp4<sup>fl/fl</sup>, and Pcp2<sup>Cre</sup>; Mito-GFP. Behavioral tests, including rotarod, open field, balance beam, and Morris water maze, were conducted. Ucp4-overexpressing virus was administered to cerebellar lobes 4/5. Mitochondrial morphology was assessed <i>via</i> transmission electron microscopy, 3D reconstruction, and network analysis, while function was evaluated through reactive oxygen species, mitochondrial membrane potential (MMP), glutathione/glutathione disulfide ratio, adenosine triphosphate levels, qPCR, and Western blotting. Results showed that IHH induces hypoactivity without affecting spatial cognition. IHH-induced hypoactivity is linked to Ucp4 upregulation and increased mitochondrial fragmentation in Purkinje cells (PCs), though overall mitochondrial dynamics remain balanced. Ucp4 deficiency exacerbates IHH-induced hypoactivity and mitochondrial fragmentation. Conversely, Ucp4 overexpression in PCs significantly alleviates these effects. Mechanistically, <i>Ucp4</i> protects PCs by stabilizing MMP and regulating oxidative stress, maintaining mitochondrial integrity. This study reveals that <i>Ucp4</i> protects cerebellar PCs from oxidative stress in IHH, improving motor function and identifying <i>Ucp4</i> as a potential therapeutic target for intermittent high-altitude syndrome. <i>Antioxid. Redox Signal.</i> 43, 483-508.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"483-508"},"PeriodicalIF":6.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144599194","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Melatonin Ameliorates Cognitive Impairment Following Exertional Heat Stroke by Inhibiting Ferroptosis and Neuroinflammation. 褪黑素通过抑制铁下垂和神经炎症改善劳累性中暑后的认知障碍。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-01 Epub Date: 2025-08-08 DOI: 10.1177/15230864251363577
Xiaochen Wang, Ziwei Han, Chao Liu, Jiaona Liu, Zhi Dai, Jie Hu, Zhi Mao, Qinglin Li, Xin Hu, Feihu Zhou

Aims: This study aims to investigate whether melatonin (MLT) exerts protective effects against cognitive impairment following exertional heat stroke (EHS) by modulating ferroportin (Fpn) to alleviate hippocampal ferroptosis and neuroinflammation. Results: Following EHS, genes such as Mt1, Mt2, and Trf were notably upregulated in the hippocampal tissue, whereas genes such as Slc40a1 (encoding Fpn 1) and Il33 were downregulated. Kyoto Encyclopedia of Genes and Genomes analysis implicated ferroptosis as a dominant. MLT significantly ameliorated learning and memory deficits observed in EHS mice. This treatment also modulated ferroptosis markers, such as Fpn, xCT, ferritin H, and glutathione peroxidase 4, reduced hippocampal iron overload, and decreased the secretion of proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-α (TNF-α). Furthermore, MLT treatment reduced oxidative stress and lipid peroxidation and mitigated mitochondrial and neuronal damage in the hippocampal tissue. Strikingly, conditional Fpn knockout abolished MLT's benefits: Fpn-cKO + MLT mice showed persistent iron accumulation, elevated IL-6 and TNF-α, and failed cognitive recovery. Innovation: Our study reveals that MLT prevents EHS-induced neurodegeneration by enhancing Fpn-dependent iron efflux, a mechanism that concurrently resolves hippocampal iron overload, suppresses ferroptosis, and dampens neuroinflammation. Conclusion: Our findings indicate that MLT mitigates EHS-related cognitive impairment by restoring hippocampal iron homeostasis and suppressing neuroinflammation, primarily through Fpn-dependent mechanisms. Antioxid. Redox Signal. 43, 509-527.

目的:探讨褪黑素(melatonin, MLT)是否通过调节铁转运蛋白(ferroportin, Fpn)减轻海马铁吊死和神经炎症,从而对运动性中暑(EHS)后认知功能障碍具有保护作用。结果:EHS后,海马组织中Mt1、Mt2、Trf等基因显著上调,而编码fpn1的Slc40a1、Il33等基因下调。京都基因和基因组百科分析表明,铁下垂是显性的。MLT显著改善了EHS小鼠的学习和记忆缺陷。这种治疗还可以调节铁下垂标志物,如Fpn、xCT、铁蛋白H和谷胱甘肽过氧化物酶4,减少海马铁超载,减少促炎细胞因子白介素(IL)-6和肿瘤坏死因子-α (TNF-α)的分泌。此外,MLT治疗减少了氧化应激和脂质过氧化,减轻了海马组织中的线粒体和神经元损伤。引人注目的是,条条性Fpn敲除消除了MLT的益处:Fpn- cko + MLT小鼠表现出持续的铁积累,IL-6和TNF-α升高,认知恢复失败。创新:我们的研究表明,MLT通过增强fpn依赖的铁外排来预防ehs诱导的神经退行性变,这一机制同时解决了海马铁过载,抑制铁下沉,并抑制神经炎症。结论:我们的研究结果表明,MLT主要通过fpn依赖机制,通过恢复海马铁稳态和抑制神经炎症来减轻ehs相关的认知障碍。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Melatonin Ameliorates Cognitive Impairment Following Exertional Heat Stroke by Inhibiting Ferroptosis and Neuroinflammation.","authors":"Xiaochen Wang, Ziwei Han, Chao Liu, Jiaona Liu, Zhi Dai, Jie Hu, Zhi Mao, Qinglin Li, Xin Hu, Feihu Zhou","doi":"10.1177/15230864251363577","DOIUrl":"10.1177/15230864251363577","url":null,"abstract":"<p><p><b><i>Aims:</i></b> This study aims to investigate whether melatonin (MLT) exerts protective effects against cognitive impairment following exertional heat stroke (EHS) by modulating ferroportin (Fpn) to alleviate hippocampal ferroptosis and neuroinflammation. <b><i>Results:</i></b> Following EHS, genes such as <i>Mt1</i>, <i>Mt2</i>, and <i>Trf</i> were notably upregulated in the hippocampal tissue, whereas genes such as <i>Slc40a1</i> (encoding Fpn 1) and <i>Il33</i> were downregulated. Kyoto Encyclopedia of Genes and Genomes analysis implicated ferroptosis as a dominant. MLT significantly ameliorated learning and memory deficits observed in EHS mice. This treatment also modulated ferroptosis markers, such as Fpn, xCT, ferritin H, and glutathione peroxidase 4, reduced hippocampal iron overload, and decreased the secretion of proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-α (TNF-α). Furthermore, MLT treatment reduced oxidative stress and lipid peroxidation and mitigated mitochondrial and neuronal damage in the hippocampal tissue. Strikingly, conditional Fpn knockout abolished MLT's benefits: Fpn-cKO + MLT mice showed persistent iron accumulation, elevated IL-6 and TNF-α, and failed cognitive recovery. <b><i>Innovation:</i></b> Our study reveals that MLT prevents EHS-induced neurodegeneration by enhancing Fpn-dependent iron efflux, a mechanism that concurrently resolves hippocampal iron overload, suppresses ferroptosis, and dampens neuroinflammation. <b><i>Conclusion:</i></b> Our findings indicate that MLT mitigates EHS-related cognitive impairment by restoring hippocampal iron homeostasis and suppressing neuroinflammation, primarily through Fpn-dependent mechanisms. <i>Antioxid. Redox Signal.</i> 43, 509-527.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"509-527"},"PeriodicalIF":6.1,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144803274","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epstein-Barr Virus Hijacks Redox Signaling via Glutathione Peroxidase 4 to Sustain Latency and Drive Gastric Cancer Progression. eb病毒通过谷胱甘肽过氧化物酶4劫持氧化还原信号以维持潜伏并驱动胃癌进展。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-30 DOI: 10.1177/15230864251382885
Duo Shi, Yanhong Zhao, Xia Zhao, Zhiyuan Gong, Wen Liu, Ping Li, Yan Zhang, Bing Luo

Aims: Epstein-Barr virus (EBV)-associated gastric cancer (GC) accounts for about 9% of GC patients, but its pathogenesis remains unclear. Glutathione peroxidase 4 (GPX4) is an important antioxidant enzyme that is highly expressed in various tumors and is associated with viral infections. This study aimed to clarify the relationship between EBV and GPX4 and the role of GPX4 in the occurrence and development of EBV-associated GC. Results: EBV infection leads to oxidative stress and excessive generation of reactive oxygen species (ROS) in GC cells. At the same time, EBV upregulates the expression of antioxidant enzyme GPX4 through the latent membrane protein 2A (LMP2A)/p62/Kelch-like ECH-associated protein 1(Keap1)/nuclear factor (erythroid-derived 2)-like 2 (NRF2) axis, eliminating excessive ROS to balance redox homeostasis and maintain its own survival. The high expression of GPX4 in GC inhibits EBV's immediate early lytic gene BZLF1 expression, thereby inhibiting EBV reactivation, and promotes cell migration and proliferation by upregulating lipocalin-2 (LCN2). Innovation: This study is the first to demonstrate that EBV-induced GPX4 expression via the LMP2A/p62/Keap1/NRF2 axis contributes to both viral latency and tumor progression in GC. Conclusion: EBV activates the p62/Keap1/NRF2 signaling pathway through LMP2A to upregulate the expression of GPX4, thereby alleviating oxidative stress caused by viral infection and maintaining the redox homeostasis in GC cells. Such enhanced expression not only maintains the latent infection of EBV but also promotes the malignant transformation of GC cells through LCN2. Antioxid. Redox Signal. 00, 000-000.

目的:eb病毒相关胃癌(Epstein-Barr virus -associated gastric cancer, GC)约占胃癌患者的9%,其发病机制尚不清楚。谷胱甘肽过氧化物酶4 (Glutathione peroxidase 4, GPX4)是一种重要的抗氧化酶,在多种肿瘤中高表达,并与病毒感染有关。本研究旨在阐明EBV与GPX4的关系,以及GPX4在EBV相关性GC发生发展中的作用。结果:EBV感染导致GC细胞氧化应激和活性氧(ROS)过量产生。同时,EBV通过潜伏膜蛋白2A (LMP2A)/p62/ kelch样ECH-associated protein 1(Keap1)/核因子(erythrod -derived 2)-like 2 (NRF2)轴上调抗氧化酶GPX4的表达,消除过多的ROS,平衡氧化还原稳态,维持自身存活。GPX4在GC中的高表达可抑制EBV的即时早期裂解基因BZLF1的表达,从而抑制EBV的再激活,并通过上调脂钙素-2 (lipocalin-2, LCN2)促进细胞迁移和增殖。创新:本研究首次证明ebv通过LMP2A/p62/Keap1/NRF2轴诱导GPX4表达有助于胃癌病毒潜伏期和肿瘤进展。结论:EBV通过LMP2A激活p62/Keap1/NRF2信号通路,上调GPX4的表达,从而缓解病毒感染引起的氧化应激,维持GC细胞氧化还原稳态。这种增强表达不仅维持EBV的潜伏感染,而且通过LCN2促进GC细胞的恶性转化。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Epstein-Barr Virus Hijacks Redox Signaling via Glutathione Peroxidase 4 to Sustain Latency and Drive Gastric Cancer Progression.","authors":"Duo Shi, Yanhong Zhao, Xia Zhao, Zhiyuan Gong, Wen Liu, Ping Li, Yan Zhang, Bing Luo","doi":"10.1177/15230864251382885","DOIUrl":"https://doi.org/10.1177/15230864251382885","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Epstein-Barr virus (EBV)-associated gastric cancer (GC) accounts for about 9% of GC patients, but its pathogenesis remains unclear. Glutathione peroxidase 4 (GPX4) is an important antioxidant enzyme that is highly expressed in various tumors and is associated with viral infections. This study aimed to clarify the relationship between EBV and GPX4 and the role of GPX4 in the occurrence and development of EBV-associated GC. <b><i>Results:</i></b> EBV infection leads to oxidative stress and excessive generation of reactive oxygen species (ROS) in GC cells. At the same time, EBV upregulates the expression of antioxidant enzyme GPX4 through the latent membrane protein 2A (LMP2A)/p62/Kelch-like ECH-associated protein 1(Keap1)/nuclear factor (erythroid-derived 2)-like 2 (NRF2) axis, eliminating excessive ROS to balance redox homeostasis and maintain its own survival. The high expression of GPX4 in GC inhibits EBV's immediate early lytic gene BZLF1 expression, thereby inhibiting EBV reactivation, and promotes cell migration and proliferation by upregulating lipocalin-2 (LCN2). <b><i>Innovation:</i></b> This study is the first to demonstrate that EBV-induced GPX4 expression <i>via</i> the LMP2A/p62/Keap1/NRF2 axis contributes to both viral latency and tumor progression in GC. <b><i>Conclusion:</i></b> EBV activates the p62/Keap1/NRF2 signaling pathway through LMP2A to upregulate the expression of GPX4, thereby alleviating oxidative stress caused by viral infection and maintaining the redox homeostasis in GC cells. Such enhanced expression not only maintains the latent infection of EBV but also promotes the malignant transformation of GC cells through LCN2. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145197781","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Endothelial Gasdermin D Induces Mitochondrial Damage and Activates the STING Pathway in Lipopolysaccharide-Accelerated Atherosclerosis. 内皮气皮蛋白D诱导线粒体损伤并激活脂多糖加速动脉粥样硬化的STING通路。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-29 DOI: 10.1177/15230864251380286
Xiaoyue Song, Junqiang Xue, Enyong Su, Shiyao Xie, Xuelin Cheng, Peng Yu, Lili Wei, Ming Liu, Hong Jiang

Aims: Chronic inflammation is a widely acknowledged contributor to the development of atherosclerosis. Gasdermin D (GSDMD) serves as a key executor of pyroptosis in inflammatory diseases. This study aims to determine the role of endothelial GSDMD in lipopolysaccharide (LPS)-accelerated atherosclerosis and elucidate its underlying molecular mechanisms. Results: GSDMD expression was aberrantly activated in both LPS-accelerated atherosclerotic animal models and oxidized low-density lipoprotein plus LPS-treated endothelial cell models. Compared with the control, endothelial GSDMD deficiency attenuated the atherogenesis progression and vascular endothelial inflammation induced by LPS and protected against the progression of mitochondrial damage, the release of mitochondrial ROS and mitochondrial DNA, and the activation of the stimulator of interferon genes (STING) pathway both in vivo and in vitro. Mechanistically, endothelial GSDMD expression mediates mitochondrial membrane permeabilization and mitochondrial damage-associated molecular patterns release and triggers the STING pathway to aggravate atherosclerotic progression. In addition, the STING pathway activation was proved to partially reverse the effects of endothelial GSDMD deficiency both in vivo and in vitro. Moreover, the signal transducer and activator of transcription 3 was identified as a positive regulator of GSDMD expression. Innovation and Conclusion: Our findings elucidate the mechanism by which endothelial GSDMD exerts its atherogenic effects by increasing mitochondrial damage and upregulating the STING pathway in LPS-accelerated atherosclerosis. GSDMD promises to be a critical therapeutic target for atherosclerotic cardiovascular diseases. Antioxid. Redox Signal. 00, 000-000.

目的:慢性炎症是公认的动脉粥样硬化发展的促进因素。Gasdermin D (GSDMD)是炎症性疾病中热亡的关键执行者。本研究旨在确定内皮GSDMD在脂多糖(LPS)加速动脉粥样硬化中的作用,并阐明其潜在的分子机制。结果:GSDMD在lps加速动脉粥样硬化动物模型和氧化低密度脂蛋白加lps处理的内皮细胞模型中表达异常激活。与对照组相比,内皮细胞GSDMD缺乏在体内和体外均可减轻LPS诱导的动脉粥样硬化进程和血管内皮炎症,抑制线粒体损伤的进展、线粒体ROS和线粒体DNA的释放以及干扰素基因刺激因子(STING)通路的激活。在机制上,内皮细胞GSDMD表达介导线粒体膜渗透和线粒体损伤相关分子模式的释放,并触发STING通路,从而加剧动脉粥样硬化的进展。此外,在体内和体外实验中,STING通路激活被证明可以部分逆转内皮细胞GSDMD缺乏的影响。此外,转录3的信号转导和激活因子被鉴定为GSDMD表达的正调节因子。创新与结论:我们的研究结果阐明了内皮细胞GSDMD在lps加速动脉粥样硬化中通过增加线粒体损伤和上调STING通路发挥其致动脉粥样硬化作用的机制。GSDMD有望成为动脉粥样硬化性心血管疾病的关键治疗靶点。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Endothelial Gasdermin D Induces Mitochondrial Damage and Activates the STING Pathway in Lipopolysaccharide-Accelerated Atherosclerosis.","authors":"Xiaoyue Song, Junqiang Xue, Enyong Su, Shiyao Xie, Xuelin Cheng, Peng Yu, Lili Wei, Ming Liu, Hong Jiang","doi":"10.1177/15230864251380286","DOIUrl":"https://doi.org/10.1177/15230864251380286","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Chronic inflammation is a widely acknowledged contributor to the development of atherosclerosis. Gasdermin D (GSDMD) serves as a key executor of pyroptosis in inflammatory diseases. This study aims to determine the role of endothelial GSDMD in lipopolysaccharide (LPS)-accelerated atherosclerosis and elucidate its underlying molecular mechanisms. <b><i>Results:</i></b> GSDMD expression was aberrantly activated in both LPS-accelerated atherosclerotic animal models and oxidized low-density lipoprotein plus LPS-treated endothelial cell models. Compared with the control, endothelial GSDMD deficiency attenuated the atherogenesis progression and vascular endothelial inflammation induced by LPS and protected against the progression of mitochondrial damage, the release of mitochondrial ROS and mitochondrial DNA, and the activation of the stimulator of interferon genes (STING) pathway both <i>in vivo</i> and <i>in vitro</i>. Mechanistically, endothelial GSDMD expression mediates mitochondrial membrane permeabilization and mitochondrial damage-associated molecular patterns release and triggers the STING pathway to aggravate atherosclerotic progression. In addition, the STING pathway activation was proved to partially reverse the effects of endothelial GSDMD deficiency both <i>in vivo</i> and <i>in vitro</i>. Moreover, the signal transducer and activator of transcription 3 was identified as a positive regulator of GSDMD expression. <b><i>Innovation and Conclusion:</i></b> Our findings elucidate the mechanism by which endothelial GSDMD exerts its atherogenic effects by increasing mitochondrial damage and upregulating the STING pathway in LPS-accelerated atherosclerosis. GSDMD promises to be a critical therapeutic target for atherosclerotic cardiovascular diseases. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-09-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145184607","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Empagliflozin Attenuates Diabetic Cardiomyopathy via Inhibiting Cardiomyocyte Ferroptosis Through the USP7/NRF2 Signaling Pathway. 恩格列净通过USP7/NRF2信号通路抑制心肌细胞凋亡减轻糖尿病心肌病
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-26 DOI: 10.1177/15230864251377765
Min Cui, Junwei Zhang, Ziwei Wang, Xiandu Jin, Hanmo Zhang, Shengzheng Zhang, Wenjun Jia, Hao Wu, Zhi Qi, Xin Qi

Aims: Diabetic cardiomyopathy (DbCM) typically manifests as diastolic dysfunction, and treating heart failure with preserved ejection fraction (HFpEF) is challenging. Empagliflozin (Empa), a sodium-glucose cotransporter 2 inhibitor, reduces hospitalization and mortality in patients with HFpEF and the risk of DbCM. However, the underlying molecular mechanisms and the specific targets remain largely unknown. Results: Glutathione peroxidase 4 (GPX4) is a key enzyme that mitigates ferroptosis. Empa treatment improved cardiac function, upregulated GPX4 expression, and reduced ferroptosis in DbCM mice. The ferroptosis inducer erastin abolished the protective effects of Empa. Through database screening, we found that nuclear factor erythroid 2-related factor 2 (NRF2) plays an important role in ferroptosis in DbCM. NRF2 was expressed at lower levels in DbCM mice, and its expression significantly increased after Empa treatment. In NRF2-knockout mice, Empa failed to improve the cardiac function of DbCM mice, upregulate the expression of GPX4, and reduce ferroptosis. Moreover, Empa increased NRF2 levels by inhibiting ubiquitin-mediated degradation. A database search predicted that the stability of NRF2 may be regulated by ubiquitin-specific protease 7 (USP7). Immunoprecipitation assays demonstrated that USP7 interacted with NRF2 and mediated its deubiquitination, thereby stabilizing NRF2. Administration of the USP7 inhibitor P5091 abolished the effects of Empa, whereas the use of adeno-associated virus serotype 9 (AAV9)-NRF2 reversed the effects of P5091. Innovation and Conclusion: Empa attenuated cardiomyocyte ferroptosis in DbCM by stabilizing NRF2 through the USP7/NRF2/GPX4 signaling pathway. Targeting the USP7/NRF2/GPX4 pathway may represent a novel therapeutic strategy for attenuating ferroptosis in DbCM, which has clinical significance. Antioxid. Redox Signal. 00, 000-000. 2022-SYDWLL-000213.

目的:糖尿病性心肌病(DbCM)通常表现为舒张功能障碍,用保留射血分数(HFpEF)治疗心力衰竭具有挑战性。恩帕列净(Empa)是一种钠-葡萄糖共转运蛋白2抑制剂,可降低HFpEF患者的住院率和死亡率以及DbCM的风险。然而,潜在的分子机制和具体的靶点在很大程度上仍然未知。结果:谷胱甘肽过氧化物酶4 (Glutathione peroxidase 4, GPX4)是减轻铁下垂的关键酶。Empa治疗改善了DbCM小鼠的心功能,上调了GPX4的表达,并减少了铁下垂。铁下垂诱导剂擦除素消除了Empa的保护作用。通过数据库筛选,我们发现核因子红细胞2相关因子2 (NRF2)在DbCM的铁下沉中起重要作用。NRF2在DbCM小鼠中表达水平较低,经Empa处理后其表达水平显著升高。在nrf2敲除小鼠中,Empa不能改善DbCM小鼠的心功能,不能上调GPX4的表达,也不能减少铁下沉。此外,Empa通过抑制泛素介导的降解来增加NRF2水平。数据库检索预测NRF2的稳定性可能受泛素特异性蛋白酶7 (USP7)的调控。免疫沉淀实验表明USP7与NRF2相互作用并介导其去泛素化,从而稳定NRF2。施用USP7抑制剂P5091可消除Empa的作用,而使用腺相关病毒血清型9 (AAV9)-NRF2可逆转P5091的作用。创新与结论:Empa通过USP7/NRF2/GPX4信号通路稳定NRF2,减轻DbCM心肌细胞铁下垂。靶向USP7/NRF2/GPX4通路可能是一种减轻DbCM铁下垂的新治疗策略,具有临床意义。Antioxid。氧化还原信号:00000 - 00000。2022 - sydwll - 000213。
{"title":"Empagliflozin Attenuates Diabetic Cardiomyopathy via Inhibiting Cardiomyocyte Ferroptosis Through the USP7/NRF2 Signaling Pathway.","authors":"Min Cui, Junwei Zhang, Ziwei Wang, Xiandu Jin, Hanmo Zhang, Shengzheng Zhang, Wenjun Jia, Hao Wu, Zhi Qi, Xin Qi","doi":"10.1177/15230864251377765","DOIUrl":"https://doi.org/10.1177/15230864251377765","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Diabetic cardiomyopathy (DbCM) typically manifests as diastolic dysfunction, and treating heart failure with preserved ejection fraction (HFpEF) is challenging. Empagliflozin (Empa), a sodium-glucose cotransporter 2 inhibitor, reduces hospitalization and mortality in patients with HFpEF and the risk of DbCM. However, the underlying molecular mechanisms and the specific targets remain largely unknown. <b><i>Results:</i></b> Glutathione peroxidase 4 (GPX4) is a key enzyme that mitigates ferroptosis. Empa treatment improved cardiac function, upregulated GPX4 expression, and reduced ferroptosis in DbCM mice. The ferroptosis inducer erastin abolished the protective effects of Empa. Through database screening, we found that nuclear factor erythroid 2-related factor 2 (NRF2) plays an important role in ferroptosis in DbCM. NRF2 was expressed at lower levels in DbCM mice, and its expression significantly increased after Empa treatment. In NRF2-knockout mice, Empa failed to improve the cardiac function of DbCM mice, upregulate the expression of GPX4, and reduce ferroptosis. Moreover, Empa increased NRF2 levels by inhibiting ubiquitin-mediated degradation. A database search predicted that the stability of NRF2 may be regulated by ubiquitin-specific protease 7 (USP7). Immunoprecipitation assays demonstrated that USP7 interacted with NRF2 and mediated its deubiquitination, thereby stabilizing NRF2. Administration of the USP7 inhibitor P5091 abolished the effects of Empa, whereas the use of adeno-associated virus serotype 9 (AAV9)-NRF2 reversed the effects of P5091. <b><i>Innovation and Conclusion:</i></b> Empa attenuated cardiomyocyte ferroptosis in DbCM by stabilizing NRF2 through the USP7/NRF2/GPX4 signaling pathway. Targeting the USP7/NRF2/GPX4 pathway may represent a novel therapeutic strategy for attenuating ferroptosis in DbCM, which has clinical significance. <i>Antioxid. Redox Signal.</i> 00, 000-000. 2022-SYDWLL-000213.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145147524","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Antioxidants & redox signaling
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1