首页 > 最新文献

Antioxidants & redox signaling最新文献

英文 中文
α-Tocopherol Ameliorates Liver Fibrosis by Inhibiting Hepatic Stellate Cell Activation by Promoting Nrf2 Nuclear Translocation. α-生育酚通过促进Nrf2核易位抑制肝星状细胞活化改善肝纤维化。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-01 Epub Date: 2025-08-01 DOI: 10.1177/15230864251364900
Rui Fang, Xue Wang, Han Zhang, Xiaolin Xie, Huan Chen, Wenting Lu, Si Zhao, Tianming Zhao, Zihao Cai, Ming Zhang, Bing Xu, Yuzheng Zhuge, Feng Zhang

Aims: α-Tocopherol is a potent natural antioxidant with a variety of biological functions and is widely used in clinical practice. However, the effect and mechanism of α-tocopherol on liver fibrosis remain unknown. The core of liver fibrosis is the activation of hepatic stellate cell (HSC). Inhibiting HSC activation may be the underlying mechanism by which α-tocopherol alleviates liver fibrosis. Results: Our study revealed that α-tocopherol improved liver injury and fibrosis in both CCl4 and bile duct ligation induced liver fibrosis model mice. α-Tocopherol inhibited HSC activation by promoting nuclear erythroid 2-related factor 2 (Nrf2) translocation into the nucleus. α-Tocopherol directly promoted Nrf2 nuclear translocation by reducing its degradation, additionally, α-tocopherol suppressed autophagy by inhibiting endoplasmic reticulum stress, resulting in increased SQSTM1 competition to bind KEAP1 and indirectly promoting Nrf2 translocation into the nucleus. The increased Nrf2 nuclear translocation upregulated the expression of antioxidant genes, thereby reducing ROS and subsequently inhibiting HSC activation. Moreover, the antifibrotic and hepatoprotective effects of α-tocopherol were verified by the addition of the Nrf2 activator-curcumin, the autophagy inhibitor-3-methyladenine and the endoplasmic reticulum stress inhibitor-sodium 4-phenylbutyrate. Innovation and Conclusion: Our study is the first to identify the mechanism by which α-tocopherol alleviates liver fibrosis. Broadly speaking, this study demonstrated that α-tocopherol promotes Nrf2 nuclear translocation by reducing Nrf2 degradation and inhibiting endoplasmic reticulum stress, which then inhibits HSC activation and ultimately ameliorates liver injury and fibrosis. Therefore, α-tocopherol may become a novel therapeutic strategy for liver fibrosis. Antioxid. Redox Signal. 43, 833-848.

目的:α-生育酚是一种有效的天然抗氧化剂,具有多种生物学功能,广泛应用于临床。然而,α-生育酚在肝纤维化中的作用和机制尚不清楚。肝纤维化的核心是肝星状细胞的活化。抑制HSC活化可能是α-生育酚减轻肝纤维化的潜在机制。结果:α-生育酚可改善CCl4和胆管结扎所致肝纤维化模型小鼠的肝损伤和纤维化。α-生育酚通过促进细胞核内红细胞2相关因子2 (Nrf2)易位抑制HSC活化。α-生育酚通过减少Nrf2降解直接促进Nrf2核易位,α-生育酚通过抑制内质网应激抑制自噬,导致SQSTM1竞争结合KEAP1,间接促进Nrf2易位入核。Nrf2核易位的增加上调了抗氧化基因的表达,从而减少了ROS,从而抑制了HSC的活化。此外,α-生育酚通过加入Nrf2激活剂姜黄素、自噬抑制剂3-甲基腺嘌呤和内质网应激抑制剂4-苯基丁酸钠,证实了α-生育酚的抗纤维化和肝保护作用。创新与结论:本研究首次明确了α-生育酚缓解肝纤维化的机制。总的来说,本研究表明α-生育酚通过减少Nrf2降解和抑制内质网应激促进Nrf2核易位,进而抑制HSC活化,最终改善肝损伤和纤维化。因此,α-生育酚可能成为治疗肝纤维化的新策略。Antioxid。氧化还原信号:00000 - 00000。
{"title":"α-Tocopherol Ameliorates Liver Fibrosis by Inhibiting Hepatic Stellate Cell Activation by Promoting Nrf2 Nuclear Translocation.","authors":"Rui Fang, Xue Wang, Han Zhang, Xiaolin Xie, Huan Chen, Wenting Lu, Si Zhao, Tianming Zhao, Zihao Cai, Ming Zhang, Bing Xu, Yuzheng Zhuge, Feng Zhang","doi":"10.1177/15230864251364900","DOIUrl":"10.1177/15230864251364900","url":null,"abstract":"<p><p><b><i>Aims:</i></b> α-Tocopherol is a potent natural antioxidant with a variety of biological functions and is widely used in clinical practice. However, the effect and mechanism of α-tocopherol on liver fibrosis remain unknown. The core of liver fibrosis is the activation of hepatic stellate cell (HSC). Inhibiting HSC activation may be the underlying mechanism by which α-tocopherol alleviates liver fibrosis. <b><i>Results:</i></b> Our study revealed that α-tocopherol improved liver injury and fibrosis in both CCl<sub>4</sub> and bile duct ligation induced liver fibrosis model mice. α-Tocopherol inhibited HSC activation by promoting nuclear erythroid 2-related factor 2 (Nrf2) translocation into the nucleus. α-Tocopherol directly promoted Nrf2 nuclear translocation by reducing its degradation, additionally, α-tocopherol suppressed autophagy by inhibiting endoplasmic reticulum stress, resulting in increased SQSTM1 competition to bind KEAP1 and indirectly promoting Nrf2 translocation into the nucleus. The increased Nrf2 nuclear translocation upregulated the expression of antioxidant genes, thereby reducing ROS and subsequently inhibiting HSC activation. Moreover, the antifibrotic and hepatoprotective effects of α-tocopherol were verified by the addition of the Nrf2 activator-curcumin, the autophagy inhibitor-3-methyladenine and the endoplasmic reticulum stress inhibitor-sodium 4-phenylbutyrate. <b><i>Innovation and Conclusion:</i></b> Our study is the first to identify the mechanism by which α-tocopherol alleviates liver fibrosis. Broadly speaking, this study demonstrated that α-tocopherol promotes Nrf2 nuclear translocation by reducing Nrf2 degradation and inhibiting endoplasmic reticulum stress, which then inhibits HSC activation and ultimately ameliorates liver injury and fibrosis. Therefore, α-tocopherol may become a novel therapeutic strategy for liver fibrosis. <i>Antioxid. Redox Signal.</i> 43, 833-848.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"833-848"},"PeriodicalIF":6.1,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144783326","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
Retinoic Acid Receptor-related Orphan Receptor α Drives Glucose Reprogramming and Mitochondrial Rescue Mitigate Subarachnoid Hemorrhage-Induced Early Brain Injury. 视黄酸受体相关孤儿受体α驱动葡萄糖重编程和线粒体救援减轻蛛网膜下腔出血诱导的早期脑损伤
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-01 DOI: 10.1177/15230864251399609
Jie Li, Jiaqi Wang, Sijing Guo, Fengchen Zhang, Yichao Jin, Xiaohua Zhang

Aims: Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular event characterized by early brain injury (EBI) within 72 h that is driven by oxidative stress, mitochondrial dysfunction, and metabolic collapse. The retinoic acid receptor-related orphan receptor alpha (RORα) is a nuclear receptor implicated in metabolic and inflammatory regulation, but it has not been studied in SAH. We aimed to determine whether RORα confers neuroprotection after SAH and to elucidate its underlying mechanisms. Methods and Results: We used mouse SAH models and primary cortical neurons to assess the RORα expression, functional outcomes, and metabolic changes. The RORα expression was markedly reduced post-SAH. Genetic knockdown or deficiency (staggerer mice) exacerbated neuronal apoptosis, neuroinflammation, and behavioral deficits. Conversely, pharmacological activation with SR1078 significantly improved neurological scores, preserved neuronal morphology, and reduced oxidative stress. RORα overexpression or SR1078 treatment enhanced neuronal viability in vitro under hemoglobin-induced stress. Transcriptomic and epigenomic profiling revealed that RORα directly regulated glucose-6-phosphate dehydrogenase and α subunit of peroxisome proliferator activated receptor-γ coactivator-1. This promoted pentose phosphate pathway flux and mitochondrial biogenesis. A metabolic flux analysis confirmed increased nicotinamide adenine dinucleotide phosphate hydrogen and glutathione synthesis, reduced reactive oxygen species accumulation, and an improved oxygen consumption rate and spare respiratory capacity. All of these results indicated a shift toward oxidative phosphorylation and enhanced bioenergetics. Innovation and Conclusion: We are the first to demonstrate that RORα activation reprogrammed neuronal glucose metabolism and strengthened antioxidant defenses to mitigate SAH-induced EBI. The targeting of RORα could represent a promising therapeutic strategy for stroke-related metabolic failure and oxidative stress. Future work should explore the translational potential in clinical settings. Antioxid. Redox Signal. 00, 000-000.

目的:蛛网膜下腔出血(SAH)是一种以72小时内早期脑损伤(EBI)为特征的破坏性脑血管事件,由氧化应激、线粒体功能障碍和代谢崩溃驱动。视黄酸受体相关孤儿受体α (RORα)是参与代谢和炎症调节的核受体,但尚未在SAH中进行研究。我们的目的是确定RORα是否在SAH后提供神经保护并阐明其潜在机制。方法和结果:我们使用小鼠SAH模型和原代皮质神经元来评估rora的表达、功能结局和代谢变化。sah后RORα表达明显降低。基因敲低或缺失(交错小鼠)加剧了神经元凋亡、神经炎症和行为缺陷。相反,SR1078的药理激活可显著改善神经学评分,保存神经元形态,减少氧化应激。RORα过表达或SR1078处理可提高血红蛋白诱导应激下的体外神经元活力。转录组学和表观基因组分析显示,rora直接调节葡萄糖-6-磷酸脱氢酶和过氧化物酶体增殖物激活受体-γ共激活因子-1的α亚基。这促进了戊糖磷酸途径通量和线粒体的生物发生。代谢通量分析证实烟酰胺腺嘌呤二核苷酸磷酸氢和谷胱甘肽合成增加,活性氧积累减少,耗氧量和备用呼吸能力提高。所有这些结果都表明了向氧化磷酸化和增强生物能量的转变。创新与结论:我们首次证明了rora激活可重编程神经元葡萄糖代谢并增强抗氧化防御以减轻sah诱导的EBI。靶向RORα可能是治疗脑卒中相关代谢衰竭和氧化应激的一种有前景的治疗策略。未来的工作应该探索在临床环境中的转化潜力。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Retinoic Acid Receptor-related Orphan Receptor α Drives Glucose Reprogramming and Mitochondrial Rescue Mitigate Subarachnoid Hemorrhage-Induced Early Brain Injury.","authors":"Jie Li, Jiaqi Wang, Sijing Guo, Fengchen Zhang, Yichao Jin, Xiaohua Zhang","doi":"10.1177/15230864251399609","DOIUrl":"https://doi.org/10.1177/15230864251399609","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Subarachnoid hemorrhage (SAH) is a devastating cerebrovascular event characterized by early brain injury (EBI) within 72 h that is driven by oxidative stress, mitochondrial dysfunction, and metabolic collapse. The retinoic acid receptor-related orphan receptor alpha (RORα) is a nuclear receptor implicated in metabolic and inflammatory regulation, but it has not been studied in SAH. We aimed to determine whether RORα confers neuroprotection after SAH and to elucidate its underlying mechanisms. <b><i>Methods and Results:</i></b> We used mouse SAH models and primary cortical neurons to assess the RORα expression, functional outcomes, and metabolic changes. The RORα expression was markedly reduced post-SAH. Genetic knockdown or deficiency (staggerer mice) exacerbated neuronal apoptosis, neuroinflammation, and behavioral deficits. Conversely, pharmacological activation with SR1078 significantly improved neurological scores, preserved neuronal morphology, and reduced oxidative stress. RORα overexpression or SR1078 treatment enhanced neuronal viability <i>in vitro</i> under hemoglobin-induced stress. Transcriptomic and epigenomic profiling revealed that RORα directly regulated glucose-6-phosphate dehydrogenase and α subunit of peroxisome proliferator activated receptor-γ coactivator-1. This promoted pentose phosphate pathway flux and mitochondrial biogenesis. A metabolic flux analysis confirmed increased nicotinamide adenine dinucleotide phosphate hydrogen and glutathione synthesis, reduced reactive oxygen species accumulation, and an improved oxygen consumption rate and spare respiratory capacity. All of these results indicated a shift toward oxidative phosphorylation and enhanced bioenergetics. <b><i>Innovation and Conclusion:</i></b> We are the first to demonstrate that RORα activation reprogrammed neuronal glucose metabolism and strengthened antioxidant defenses to mitigate SAH-induced EBI. The targeting of RORα could represent a promising therapeutic strategy for stroke-related metabolic failure and oxidative stress. Future work should explore the translational potential in clinical settings. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145666814","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
SIRT3-FOXO3a Isoforms Forge Nuclear-Mitochondrial Links to Combat Sepsis-Induced Cardiomyopathy Oxidative Stress in Mice. SIRT3-FOXO3a同种异构体形成核线粒体链接以对抗败血症诱导的小鼠心肌病氧化应激
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-01 Epub Date: 2025-09-10 DOI: 10.1177/15230864251374227
Xun Luo, Zhengguang Geng, Han Zhang, Wenbo Chen, Junwen Zhang, Siyi Ming, Shiyuan Wang, Mingchun Wang, Haiyun Lei, Bao Fu, Xiaoyun Fu

Aims: Sepsis-induced cardiomyopathy (SIC) is a serious complication of sepsis. The relationship between SIC and protein acetylation, particularly the balance between acetylation and deacetylation in cardiomyocyte subcellular structures, as well as how nuclear-mitochondrial coordination maintains standard antioxidant stress capacity, remains unclear. This study focused on exploring the nuclear-mitochondrial regulatory mechanisms formed by the interplay of Sirtuin 3 (SIRT3) and Forkhead box O3a (FOXO3a). Results: In vivo, SIC markers increased significantly in wild-type CLP (Cecal Ligation and Puncture) mice at 72 h (CLP72h) but were partially reversed in CLP72h+oeSIRT3 mice. CLP72h mice exhibited significantly reduced mitochondrial area, aspect ratio, and mtDNA copy number. Echocardiography revealed significantly impaired cardiac function. Western blotting showed significantly decreased nuclear and mitochondrial long-form SIRT3, nuclear long-form and mitochondrial short-form FOXO3a, and mitochondrial superoxide dismutase 2 (SOD2), with significantly increased acetylation in CLP72h mice. In vitro, oeSIRT3 preserved nuclear FOXO3a localization and mitochondrial membrane potential, with CLP72h+oeSIRT3 mice showing significantly reduced oxidative stress. The long form of SIRT3 plays a crucial deacetylation role in SIC and influences SOD2 partially through FOXO3a. Innovation: This study explored the roles of different SIRT3 and FOXO3a isoforms in combating oxidative stress in SIC through dynamic nucleus-mitochondrial regulation. Conclusion: This study underscores the critical role of the SIRT3-FOXO3a axis in enhancing mitochondrial antioxidant capacity through a nuclear-mitochondrial network during SIC, offering new insights into molecular mechanisms and potential therapeutic strategies for SIC. Antioxid. Redox Signal. 43, 805-818.

目的:败血症性心肌病(SIC)是败血症的严重并发症。SIC与蛋白质乙酰化之间的关系,特别是心肌细胞亚细胞结构中乙酰化和去乙酰化之间的平衡,以及核-线粒体协调如何维持标准的抗氧化应激能力,目前尚不清楚。本研究重点探讨Sirtuin 3 (SIRT3)与叉头盒O3a (FOXO3a)相互作用形成的核线粒体调控机制。结果:在体内,野生型CLP(盲肠结扎和穿刺)小鼠的SIC标记在72h (CLP72h)时显著升高,而在CLP72h+oeSIRT3小鼠中部分逆转。CLP72h小鼠的线粒体面积、纵横比和mtDNA拷贝数显著减少。超声心动图显示心功能明显受损。Western blotting显示,CLP72h小鼠细胞核和线粒体长型SIRT3、细胞核长型和线粒体短型FOXO3a、线粒体超氧化物歧化酶2 (SOD2)显著降低,乙酰化显著升高。在体外,oeSIRT3保留了细胞核FOXO3a的定位和线粒体膜电位,CLP72h+oeSIRT3小鼠的氧化应激显著降低。长链SIRT3在SIC中起着至关重要的去乙酰化作用,并通过FOXO3a部分影响SOD2。创新:本研究探讨了不同SIRT3和FOXO3a亚型通过核-线粒体动态调控对抗SIC氧化应激的作用。结论:本研究强调SIRT3-FOXO3a轴在SIC过程中通过核-线粒体网络增强线粒体抗氧化能力的关键作用,为SIC的分子机制和潜在的治疗策略提供了新的见解。Antioxid。氧化还原信号:00000 - 00000。
{"title":"SIRT3-FOXO3a Isoforms Forge Nuclear-Mitochondrial Links to Combat Sepsis-Induced Cardiomyopathy Oxidative Stress in Mice.","authors":"Xun Luo, Zhengguang Geng, Han Zhang, Wenbo Chen, Junwen Zhang, Siyi Ming, Shiyuan Wang, Mingchun Wang, Haiyun Lei, Bao Fu, Xiaoyun Fu","doi":"10.1177/15230864251374227","DOIUrl":"10.1177/15230864251374227","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Sepsis-induced cardiomyopathy (SIC) is a serious complication of sepsis. The relationship between SIC and protein acetylation, particularly the balance between acetylation and deacetylation in cardiomyocyte subcellular structures, as well as how nuclear-mitochondrial coordination maintains standard antioxidant stress capacity, remains unclear. This study focused on exploring the nuclear-mitochondrial regulatory mechanisms formed by the interplay of Sirtuin 3 (SIRT3) and Forkhead box O3a (FOXO3a). <b><i>Results:</i></b> <i>In vivo</i>, SIC markers increased significantly in wild-type CLP (Cecal Ligation and Puncture) mice at 72 h (CLP72h) but were partially reversed in CLP72h+oeSIRT3 mice. CLP72h mice exhibited significantly reduced mitochondrial area, aspect ratio, and mtDNA copy number. Echocardiography revealed significantly impaired cardiac function. Western blotting showed significantly decreased nuclear and mitochondrial long-form SIRT3, nuclear long-form and mitochondrial short-form FOXO3a, and mitochondrial superoxide dismutase 2 (SOD2), with significantly increased acetylation in CLP72h mice. <i>In vitro</i>, oeSIRT3 preserved nuclear FOXO3a localization and mitochondrial membrane potential, with CLP72h+oeSIRT3 mice showing significantly reduced oxidative stress. The long form of SIRT3 plays a crucial deacetylation role in SIC and influences SOD2 partially through FOXO3a. <b><i>Innovation:</i></b> This study explored the roles of different SIRT3 and FOXO3a isoforms in combating oxidative stress in SIC through dynamic nucleus-mitochondrial regulation. <b><i>Conclusion:</i></b> This study underscores the critical role of the SIRT3-FOXO3a axis in enhancing mitochondrial antioxidant capacity through a nuclear-mitochondrial network during SIC, offering new insights into molecular mechanisms and potential therapeutic strategies for SIC. <i>Antioxid. Redox Signal.</i> 43, 805-818.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"805-818"},"PeriodicalIF":6.1,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145032612","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
Nimodipine Blocks Histone-Induced Calcium Overload to Protect Neurons after Traumatic Brain Injury. 尼莫地平阻断组蛋白诱导的钙超载对脑外伤后神经元的保护作用。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-01 Epub Date: 2025-09-17 DOI: 10.1177/15230864251376030
Wei Cao, Yunfeng Xu

Aims: To investigate if nimodipine alleviates traumatic brain injury (TBI)-induced neuronal apoptosis and neurological deficits by inhibiting extracellular histone-mediated Ca2+ influx, mitochondrial damage, and Caspase pathway activation. Results: In vitro, nimodipine significantly reduced histone-induced Ca2+ influx in cortical neurons, reversed by Ca2+ activator A23187. It restored neuronal proliferation (↑3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, ↑Ki67+ cells), reduced apoptosis (↓Annexin V/propidium iodide), improved mitochondrial function (↑ΔΨm/adenosine triphosphate, ↓reactive oxygen species/malondialdehyde, ↑Glutathione Peroxidase), and modulated apoptosis markers (↓Bax, ↑Bcl-2). These effects were blocked by A23187 or Caspase activator AD-2646, which increased Cleaved Caspase-3/9 and PARP1. Molecular docking confirmed nimodipine-histone binding. Transcriptomics revealed nimodipine reversed histone-induced dysregulation of Ca2+ signaling, mitochondrial apoptosis, and oxidative stress pathways, with Caspase-3 as a key protein-protein interaction node. In vivo, nimodipine improved spatial memory (Morris maze), neurological function (↓modified neurological severity score), and motor coordination (↑rotarod) in TBI mice. It reduced brain lesions (2,3,5-triphenyltetrazolium chloride), neuronal loss (hematoxylin and eosin/Nissl), Ca2+ accumulation, and proapoptotic protein expression and restored ΔΨm. Histone coadministration attenuated these benefits. Innovation: First demonstration that nimodipine directly targets extracellular histone-induced Ca2+ influx-a key TBI pathology mechanism-preserving mitochondrial integrity and inhibiting the Caspase cascade, extending beyond its known vasodilatory effects. Conclusion: Nimodipine mitigates post-TBI neuronal apoptosis and dysfunction by blocking extracellular histone-driven Ca2+ overload, preventing mitochondrial damage, and suppressing Caspase activation, significantly improving functional recovery. Antioxid. Redox Signal. 43, 869-885.

目的:研究尼莫地平是否通过抑制细胞外组蛋白介导的Ca2+内流、线粒体损伤和Caspase通路激活来缓解创伤性脑损伤(TBI)诱导的神经元凋亡和神经功能缺损。结果:在体外,尼莫地平显著减少组蛋白诱导的皮层神经元Ca2+内流,Ca2+激活剂A23187逆转。它恢复了神经元的增殖(↑3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四唑,↑Ki67+细胞),减少了细胞凋亡(↓Annexin V/碘化丙啶),改善了线粒体功能(↑ΔΨm/三磷酸腺苷,↓活性氧/丙二醛,↑谷胱甘肽过氧化物酶),并调节了细胞凋亡标志物(↓Bax,↑Bcl-2)。这些作用被A23187或Caspase激活剂AD-2646阻断,从而增加了裂解的Caspase-3/9和PARP1。分子对接证实尼莫地平与组蛋白结合。转录组学显示尼莫地平逆转组蛋白诱导的Ca2+信号失调、线粒体凋亡和氧化应激途径,其中Caspase-3是关键的蛋白相互作用节点。在体内,尼莫地平改善了TBI小鼠的空间记忆(Morris迷宫)、神经功能(↓修改神经严重程度评分)和运动协调(↑rotarod)。它减少了脑损伤(2,3,5-三苯四唑氯),神经元损失(苏木精和伊红/Nissl), Ca2+积累和促凋亡蛋白表达,并恢复ΔΨm。组蛋白联合用药减弱了这些益处。创新:首次证明尼莫地平直接靶向细胞外组蛋白诱导的Ca2+流-一个关键的TBI病理机制-保持线粒体完整性和抑制Caspase级联,扩展超出其已知的血管舒张作用。结论:尼莫地平通过阻断细胞外组蛋白驱动的Ca2+超载,防止线粒体损伤,抑制Caspase激活,显著改善脑外伤后神经元的凋亡和功能障碍,显著改善功能恢复。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Nimodipine Blocks Histone-Induced Calcium Overload to Protect Neurons after Traumatic Brain Injury.","authors":"Wei Cao, Yunfeng Xu","doi":"10.1177/15230864251376030","DOIUrl":"10.1177/15230864251376030","url":null,"abstract":"<p><p><b><i>Aims:</i></b> To investigate if nimodipine alleviates traumatic brain injury (TBI)-induced neuronal apoptosis and neurological deficits by inhibiting extracellular histone-mediated Ca<sup>2+</sup> influx, mitochondrial damage, and Caspase pathway activation. <b><i>Results:</i></b> In vitro, nimodipine significantly reduced histone-induced Ca<sup>2+</sup> influx in cortical neurons, reversed by Ca<sup>2+</sup> activator A23187. It restored neuronal proliferation (↑3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, ↑Ki67+ cells), reduced apoptosis (↓Annexin V/propidium iodide), improved mitochondrial function (↑ΔΨm/adenosine triphosphate, ↓reactive oxygen species/malondialdehyde, ↑Glutathione Peroxidase), and modulated apoptosis markers (↓Bax, ↑Bcl-2). These effects were blocked by A23187 or Caspase activator AD-2646, which increased Cleaved Caspase-3/9 and PARP1. Molecular docking confirmed nimodipine-histone binding. Transcriptomics revealed nimodipine reversed histone-induced dysregulation of Ca<sup>2+</sup> signaling, mitochondrial apoptosis, and oxidative stress pathways, with Caspase-3 as a key protein-protein interaction node. In vivo, nimodipine improved spatial memory (Morris maze), neurological function (↓modified neurological severity score), and motor coordination (↑rotarod) in TBI mice. It reduced brain lesions (2,3,5-triphenyltetrazolium chloride), neuronal loss (hematoxylin and eosin/Nissl), Ca<sup>2+</sup> accumulation, and proapoptotic protein expression and restored ΔΨm. Histone coadministration attenuated these benefits. <b><i>Innovation:</i></b> First demonstration that nimodipine directly targets extracellular histone-induced Ca<sup>2+</sup> influx-a key TBI pathology mechanism-preserving mitochondrial integrity and inhibiting the Caspase cascade, extending beyond its known vasodilatory effects. <b><i>Conclusion:</i></b> Nimodipine mitigates post-TBI neuronal apoptosis and dysfunction by blocking extracellular histone-driven Ca<sup>2+</sup> overload, preventing mitochondrial damage, and suppressing Caspase activation, significantly improving functional recovery. <i>Antioxid. Redox Signal.</i> 43, 869-885.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"869-885"},"PeriodicalIF":6.1,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145079551","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
THRA Orchestrates Myocardial Protection Against Hypothyroidism-Induced Ferroptosis via the GATA4-GPX4 Transcriptional Cascade. THRA通过GATA4-GPX4转录级联调控心肌保护以对抗甲状腺功能减退症诱导的铁下垂。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-26 DOI: 10.1177/15230864251399166
Zhang Runfang, Xianen Fa

Aims: Hypothyroidism frequently causes myocardial injury, but the role of thyroid hormone receptor alpha (THRA) remains unclear. This study investigated the function and mechanism of THRA in hypothyroidism-associated cardiac damage. Methods: A propylthiouracil (PTU)-induced hypothyroid mouse model was utilized, incorporating wild-type and THRA-knockout (KO) groups with or without thyroxine (T4) treatment. Systemic parameters, cardiac injury, histopathology, and molecular pathways were analyzed using enzyme-linked immunosorbent assay, immunohistochemistry, Western blot, quantitative polymerase chain reaction, RNA sequencing, chromatin immunoprecipitation, and dual-luciferase reporter assays. Results: PTU-induced hypothyroidism significantly reduced body weight, impaired cardiac function, and dysregulated thyroid hormones. THRA KO exacerbated these effects and completely abolished the therapeutic response to T4. Crucially, group KO-M markedly elevated markers of ferroptosis, including iron overload, malondialdehyde, and reactive oxygen species, while suppressing the reduced-to-oxidized glutathione ratio (GSH/GSSG) and key antiferroptotic proteins like glutathione peroxidase 4 (GPX4), compared with group M. Mechanistically, we identified GATA binding protein 4 (GATA4) as an upstream transcriptional activator of THRA. Furthermore, THRA itself directly bound to the GPX4 promoter and transactivated its expression. This GATA4-THRA-GPX4 axis was essential for cardioprotection, alongside modulation of the phosphoinositide 3-kinase/protein kinase B signaling pathway. Conclusion: This study defines the GATA4-THRA-GPX4 transcriptional axis as a crucial mechanism that protects the heart from hypothyroidism-driven ferroptosis, uncovering a previously unrecognized transcriptional axis that is crucial for cardioprotection during hypothyroidism. Antioxid. Redox Signal. 00, 000-000.

目的:甲状腺功能减退常引起心肌损伤,但甲状腺激素受体α (THRA)的作用尚不清楚。本研究探讨THRA在甲状腺功能减退相关心脏损伤中的作用及机制。方法:采用丙基硫脲嘧啶(PTU)诱导的甲状腺功能减退小鼠模型,分为甲状腺素(T4)治疗的野生型和thra敲除(KO)组。采用酶联免疫吸附试验、免疫组织化学、Western blot、定量聚合酶链反应、RNA测序、染色质免疫沉淀和双荧光素酶报告基因分析系统参数、心脏损伤、组织病理学和分子通路。结果:ptu诱导的甲状腺功能减退症显著降低体重,心功能受损,甲状腺激素失调。THRA KO加重了这些影响,并完全消除了对T4的治疗反应。关键是,与m组相比,KO-M组显著提高了铁超载、丙二醛和活性氧等铁死亡标志物,同时抑制了还原性氧化谷胱甘肽比率(GSH/GSSG)和关键的抗铁死亡蛋白,如谷胱甘肽过氧化物酶4 (GPX4)。此外,THRA本身直接结合GPX4启动子并反激活其表达。这个GATA4-THRA-GPX4轴对心脏保护至关重要,同时还调节磷酸肌苷3-激酶/蛋白激酶B信号通路。结论:本研究将GATA4-THRA-GPX4转录轴定义为保护心脏免受甲状腺功能减退驱动的铁上垂症的关键机制,揭示了一个以前未被认识的转录轴,该转录轴在甲状腺功能减退期间对心脏保护至关重要。Antioxid。氧化还原信号:00000 - 00000。
{"title":"THRA Orchestrates Myocardial Protection Against Hypothyroidism-Induced Ferroptosis <i>via</i> the GATA4-GPX4 Transcriptional Cascade.","authors":"Zhang Runfang, Xianen Fa","doi":"10.1177/15230864251399166","DOIUrl":"https://doi.org/10.1177/15230864251399166","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Hypothyroidism frequently causes myocardial injury, but the role of <i>thyroid hormone receptor alpha</i> (<i>THRA</i>) remains unclear. This study investigated the function and mechanism of <i>THRA</i> in hypothyroidism-associated cardiac damage. <b><i>Methods:</i></b> A propylthiouracil (PTU)-induced hypothyroid mouse model was utilized, incorporating wild-type and <i>THRA</i>-knockout (KO) groups with or without thyroxine (T4) treatment. Systemic parameters, cardiac injury, histopathology, and molecular pathways were analyzed using enzyme-linked immunosorbent assay, immunohistochemistry, Western blot, quantitative polymerase chain reaction, RNA sequencing, chromatin immunoprecipitation, and dual-luciferase reporter assays. <b><i>Results:</i></b> PTU-induced hypothyroidism significantly reduced body weight, impaired cardiac function, and dysregulated thyroid hormones. <i>THRA</i> KO exacerbated these effects and completely abolished the therapeutic response to T4. Crucially, group KO-M markedly elevated markers of ferroptosis, including iron overload, malondialdehyde, and reactive oxygen species, while suppressing the reduced-to-oxidized glutathione ratio (GSH/GSSG) and key antiferroptotic proteins like <i>glutathione peroxidase 4</i> (<i>GPX4</i>), compared with group M. Mechanistically, we identified <i>GATA binding protein 4</i> (<i>GATA4</i>) as an upstream transcriptional activator of <i>THRA</i>. Furthermore, <i>THRA</i> itself directly bound to the <i>GPX4</i> promoter and transactivated its expression. This <i>GATA4</i>-<i>THRA</i>-<i>GPX4</i> axis was essential for cardioprotection, alongside modulation of the <i>phosphoinositide 3-kinase</i>/<i>protein kinase B</i> signaling pathway. <b><i>Conclusion:</i></b> This study defines the <i>GATA4</i>-<i>THRA</i>-<i>GPX4</i> transcriptional axis as a crucial mechanism that protects the heart from hypothyroidism-driven ferroptosis, uncovering a previously unrecognized transcriptional axis that is crucial for cardioprotection during hypothyroidism. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-11-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145628121","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
Polypeptide Nanomicrospheres Delivering Dexmedetomidine Mitigate Intestinal Ischemia/Reperfusion Injury by Regulating Autophagy and Inflammation. 多肽纳米微球递送右美托咪定通过调节自噬和炎症减轻肠道缺血/再灌注损伤。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-24 DOI: 10.1177/15230864251394744
Zhen Liu, Qian Hu, Qiuhong Chen, Qiong Wu, Sisi Liang, Jinping Nie, Chenlu Fan, Qin Zhang, Xuekang Zhang

Aims: This study aims to evaluate the protective effects of dexmedetomidine-loaded polypeptide nanomicrospheres (PNM@Dex) in a mouse model of intestinal ischemia/reperfusion (I/R) injury and investigate the underlying molecular mechanisms, focusing on autophagy activation in enteric neurons. Results: PNM@Dex, synthesized via solid-phase peptide synthesis and coprecipitation, exhibited uniform spherical morphology (∼150 nm) and high drug encapsulation efficiency. In vitro studies demonstrated that PNM@Dex promoted autophagy in enteric neurons, attenuated oxidative stress and apoptosis, and improved cell viability. In vivo administration significantly mitigated intestinal injury, suppressed inflammatory cytokine production, and increased the expression of autophagy-related proteins. Autophagy inhibition assays confirmed the essential role of autophagy in mediating the protective effects of PNM@Dex. Innovation: This study represents the first successful incorporation of dexmedetomidine into polypeptide nanomicrospheres for targeted delivery to enteric neurons. The nanoplatform achieved sustained release, enhanced autophagy, and exerted strong anti-inflammatory and antioxidant effects, offering a novel therapeutic approach for intestinal I/R injury and potential protection in intestinal transplantation. Conclusion: PNM@Dex effectively alleviated intestinal I/R-induced damage through autophagy induction, oxidative stress reduction, and inflammation modulation, underscoring its promise as a therapeutic strategy for intestinal protection and transplantation. Antioxid. Redox Signal. 00, 000-000.

目的:本研究旨在评估右美托咪定负载多肽纳米微球(PNM@Dex)对小鼠肠缺血/再灌注(I/R)损伤模型的保护作用,并探讨其潜在的分子机制,重点关注肠神经元的自噬激活。结果:PNM@Dex通过固相多肽合成和共沉淀法合成,具有均匀的球形形貌(~ 150 nm)和较高的药物包封效率。体外研究表明PNM@Dex促进肠神经元自噬,减轻氧化应激和细胞凋亡,提高细胞活力。体内给药可显著减轻肠道损伤,抑制炎症细胞因子的产生,增加自噬相关蛋白的表达。自噬抑制实验证实了自噬在介导PNM@Dex保护作用中的重要作用。创新:该研究首次成功地将右美托咪定整合到多肽纳米微球中,用于靶向递送肠神经元。该纳米平台具有缓释、增强自噬、抗炎、抗氧化等功能,为肠I/R损伤的治疗提供了新的途径,并在肠移植中具有潜在的保护作用。结论:PNM@Dex可通过诱导自噬、减少氧化应激、调节炎症等方式有效缓解I/ r诱导的肠道损伤,有望成为肠道保护和移植的治疗策略。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Polypeptide Nanomicrospheres Delivering Dexmedetomidine Mitigate Intestinal Ischemia/Reperfusion Injury by Regulating Autophagy and Inflammation.","authors":"Zhen Liu, Qian Hu, Qiuhong Chen, Qiong Wu, Sisi Liang, Jinping Nie, Chenlu Fan, Qin Zhang, Xuekang Zhang","doi":"10.1177/15230864251394744","DOIUrl":"https://doi.org/10.1177/15230864251394744","url":null,"abstract":"<p><p><b><i>Aims:</i></b> This study aims to evaluate the protective effects of dexmedetomidine-loaded polypeptide nanomicrospheres (PNM@Dex) in a mouse model of intestinal ischemia/reperfusion (I/R) injury and investigate the underlying molecular mechanisms, focusing on autophagy activation in enteric neurons. <b><i>Results:</i></b> PNM@Dex, synthesized <i>via</i> solid-phase peptide synthesis and coprecipitation, exhibited uniform spherical morphology (∼150 nm) and high drug encapsulation efficiency. <i>In vitro</i> studies demonstrated that PNM@Dex promoted autophagy in enteric neurons, attenuated oxidative stress and apoptosis, and improved cell viability. <i>In vivo</i> administration significantly mitigated intestinal injury, suppressed inflammatory cytokine production, and increased the expression of autophagy-related proteins. Autophagy inhibition assays confirmed the essential role of autophagy in mediating the protective effects of PNM@Dex. <b><i>Innovation:</i></b> This study represents the first successful incorporation of dexmedetomidine into polypeptide nanomicrospheres for targeted delivery to enteric neurons. The nanoplatform achieved sustained release, enhanced autophagy, and exerted strong anti-inflammatory and antioxidant effects, offering a novel therapeutic approach for intestinal I/R injury and potential protection in intestinal transplantation. <b><i>Conclusion:</i></b> PNM@Dex effectively alleviated intestinal I/R-induced damage through autophagy induction, oxidative stress reduction, and inflammation modulation, underscoring its promise as a therapeutic strategy for intestinal protection and transplantation. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145666799","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
Ferroptosis in Tubular Epithelial Cells Across Distinct Renal Regions Is a Primary Causal Factor for Lupus Nephritis. 不同肾区小管上皮细胞的铁下垂是狼疮性肾炎的主要原因。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-21 DOI: 10.1177/15230864251396446
Wumeng Jin, Hualing Chen, Shuanglin Zhang, Xiaofen Xu, Peng Zhao, Xuanming Hu, Guanqun Xie, Yu Du, Chengping Wen, Lu Lu, Changfeng Hu

Aims: Ferroptosis has been implicated in the pathogenesis of lupus nephritis (LN), yet its precise role and mechanisms remain unclear. This study aimed to clarify the role of ferroptosis in LN progression and its underlying mechanisms. Methods: Transmission electron microscopy (TEM) was used to assess mitochondrial morphology in renal tissues from LN patients and MRL/lpr mice. Multidimensional mass spectrometry-based shotgun lipidomics was applied to analyze lipid alterations in renal cortex, medulla, and isolated renal tubules. Immunoblotting and reverse transcription quantitative PCR were performed to evaluate ferroptosis-related proteins and their messenger RNAs (mRNAs). Primary renal tubular epithelial cells (RTECs) from the distinct renal regions (cortex/medulla) were isolated and exposed to oxidative stress in vitro. Ferroptosis inducer erastin and inhibitor ferrostatin-1 (Fer-1) were used in vivo to determine causal effects. Results: TEM revealed typical ferroptotic mitochondrial changes in renal tissues from both LN patients and lupus-prone mice. In MRL/lpr mice, ferroptosis occurred as early as the pre-LN stage (8 weeks) and worsened by 14 weeks, with cortical tubules showing more severe damage than medullary tubules. Lipidomics demonstrated significant increases in lysophospholipids (e.g., 22:4 lysophosphatidylethanolamine, p < 0.001; 20:4 lysophosphatidylcholine, p < 0.01) and HNE species (p < 0.05), along with reductions in plasmalogens (e.g., 18:1-20:4 plasmenylcholine, p < 0.001). Mechanistically, ferroptosis was driven by downregulation of glutathione peroxidase 4 (p < 0.001) and solute carrier family 7 member 11 (p < 0.01) and upregulation of Acyl-CoA synthetase long chain family member 4 (p < 0.05), consistent with mRNA changes. Functionally, cortical RTECs cultured in vitro exhibited higher lipid reactive oxygen species (p < 0.001) and ferrous ion (Fe2+) accumulation (p < 0.01). In vivo, erastin accelerated LN progression, whereas Fer-1 significantly reduced proteinuria, renal pathology, and inflammatory cytokines. Innovation and Conclusion: The study provided direct evidence of ferroptosis markers in renal tissues of LN patients. RTECs exhibited the intrinsic abnormalities that trigger ferroptosis, greatly contributing to the progression of LN. Our findings highlighted the critical role of region-specific tubular ferroptosis in driving renal pathology. Early intervention targeting ferroptosis of RTECs in the renal cortex might be an effective strategy for treating LN. Antioxid. Redox Signal. 00, 000-000.

目的:上睑下垂与狼疮性肾炎(LN)的发病机制有关,但其确切的作用和机制尚不清楚。本研究旨在阐明铁下垂在LN进展中的作用及其潜在机制。方法:采用透射电镜(TEM)观察LN患者和MRL/lpr小鼠肾组织线粒体形态。基于多维质谱的散弹枪脂质组学应用于分析肾皮质、髓质和离体肾小管的脂质改变。采用免疫印迹法和反转录定量PCR法检测凋亡相关蛋白及其信使rna (mrna)。从不同的肾区(皮质/髓质)分离出原代肾小管上皮细胞(RTECs),并在体外暴露于氧化应激。在体内使用铁下垂诱导剂erastin和抑制剂ferstat -1 (fer1)来确定因果关系。结果:透射电镜显示,LN患者和狼疮易感小鼠肾组织均有典型的铁致线粒体改变。在MRL/lpr小鼠中,早在ln前期(8周)就发生了铁下垂,并在14周时恶化,皮质小管的损伤比髓小管更严重。脂质组学显示溶血磷脂(例如,22:4溶血磷脂酰乙醇胺,p < 0.001; 20:4溶血磷脂酰胆碱,p < 0.01)和HNE物种显著增加(p < 0.05),同时浆磷脂原减少(例如,18:1-20:4浆磷脂胆碱,p < 0.001)。机制上,谷胱甘肽过氧化物酶4 (p < 0.001)和溶质载体家族7成员11 (p < 0.01)下调,酰基辅酶a合成酶长链家族成员4 (p < 0.05)上调,与mRNA变化一致。在功能上,体外培养的皮质RTECs表现出较高的脂质活性氧(p < 0.001)和铁离子(Fe2+)积累(p < 0.01)。在体内,erastin加速LN进展,而Fer-1显著减少蛋白尿、肾脏病理和炎症细胞因子。创新与结论:本研究为LN患者肾组织中铁下垂标志物的存在提供了直接证据。rtec表现出触发铁下垂的内在异常,极大地促进了LN的进展。我们的研究结果强调了区域特异性小管铁下垂在驱动肾脏病理中的关键作用。针对肾皮质rtec铁下垂的早期干预可能是治疗LN的有效策略。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Ferroptosis in Tubular Epithelial Cells Across Distinct Renal Regions Is a Primary Causal Factor for Lupus Nephritis.","authors":"Wumeng Jin, Hualing Chen, Shuanglin Zhang, Xiaofen Xu, Peng Zhao, Xuanming Hu, Guanqun Xie, Yu Du, Chengping Wen, Lu Lu, Changfeng Hu","doi":"10.1177/15230864251396446","DOIUrl":"https://doi.org/10.1177/15230864251396446","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Ferroptosis has been implicated in the pathogenesis of lupus nephritis (LN), yet its precise role and mechanisms remain unclear. This study aimed to clarify the role of ferroptosis in LN progression and its underlying mechanisms. <b><i>Methods:</i></b> Transmission electron microscopy (TEM) was used to assess mitochondrial morphology in renal tissues from LN patients and MRL/<i>lpr</i> mice. Multidimensional mass spectrometry-based shotgun lipidomics was applied to analyze lipid alterations in renal cortex, medulla, and isolated renal tubules. Immunoblotting and reverse transcription quantitative PCR were performed to evaluate ferroptosis-related proteins and their messenger RNAs (mRNAs). Primary renal tubular epithelial cells (RTECs) from the distinct renal regions (cortex/medulla) were isolated and exposed to oxidative stress <i>in vitro</i>. Ferroptosis inducer erastin and inhibitor ferrostatin-1 (Fer-1) were used <i>in vivo</i> to determine causal effects. <b><i>Results:</i></b> TEM revealed typical ferroptotic mitochondrial changes in renal tissues from both LN patients and lupus-prone mice. In MRL/<i>lpr</i> mice, ferroptosis occurred as early as the pre-LN stage (8 weeks) and worsened by 14 weeks, with cortical tubules showing more severe damage than medullary tubules. Lipidomics demonstrated significant increases in lysophospholipids (<i>e.g.,</i> 22:4 lysophosphatidylethanolamine, <i>p</i> < 0.001; 20:4 lysophosphatidylcholine, <i>p</i> < 0.01) and HNE species (<i>p</i> < 0.05), along with reductions in plasmalogens (<i>e.g.,</i> 18:1-20:4 plasmenylcholine, <i>p</i> < 0.001). Mechanistically, ferroptosis was driven by downregulation of glutathione peroxidase 4 (<i>p</i> < 0.001) and solute carrier family 7 member 11 (<i>p</i> < 0.01) and upregulation of Acyl-CoA synthetase long chain family member 4 (<i>p</i> < 0.05), consistent with mRNA changes. Functionally, cortical RTECs cultured <i>in vitro</i> exhibited higher lipid reactive oxygen species (<i>p</i> < 0.001) and ferrous ion (Fe<sup>2+</sup>) accumulation (<i>p</i> < 0.01). <i>In vivo</i>, erastin accelerated LN progression, whereas Fer-1 significantly reduced proteinuria, renal pathology, and inflammatory cytokines. <b><i>Innovation and Conclusion:</i></b> The study provided direct evidence of ferroptosis markers in renal tissues of LN patients. RTECs exhibited the intrinsic abnormalities that trigger ferroptosis, greatly contributing to the progression of LN. Our findings highlighted the critical role of region-specific tubular ferroptosis in driving renal pathology. Early intervention targeting ferroptosis of RTECs in the renal cortex might be an effective strategy for treating LN. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145572734","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
Dose-Dependent Ferroptosis Induction in Osteosarcoma via NRF2/GPX4 Axis Modulation by Plasma-Activated Liquid Therapy. 血浆活化液体疗法通过NRF2/GPX4轴调节诱导骨肉瘤铁凋亡的剂量依赖性
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-20 DOI: 10.1177/15230864251399168
Longze Xiao, Mian Zhang, Peng He, Hao Wang, Sheng Li, Yonghong Wu, Shenglin Xu, Yong Hu

Aims: Plasma-activated liquid (PAL), an indirect application form of cold-atmospheric plasma (CAP)-an ionized gas generating reactive oxygen and nitrogen species, has been proposed as an innovative therapeutic approach for various cancer types. Despite accumulating evidence suggesting that PAL induces cell death through multiple mechanisms, the involvement of ferroptosis, a form of cell death driven by iron and lipid peroxidation, in osteosarcoma (OS) remains predominantly unknown. Results: CAP was used to activate the liquid for various durations, resulting in different doses of PAL. The antitumor efficacy of PAL was directly correlated with both the dosage and duration of treatment and was achieved by increasing the level of intracellular reactive oxygen species. Through screening three effective PAL doses, we discovered that PAL significantly influenced the migration and invasion capabilities of OS cells. Proteomic sequencing revealed increases in several ferroptosis-related antioxidant proteins in the PAL-treated group. Subsequent findings revealed that PAL modulated nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream ferroptosis-related genes, predominantly resulting in the induction of ferroptosis by depleting glutathione peroxidase 4 (GPX4) in human OS cells. Finally, utilizing an OS xenograft model, we found that PAL effectively suppressed tumor growth in vivo via ferroptosis. Innovation: Our study highlights the importance of the NRF2/GPX4 axis as a pivotal pathway in PAL-induced ferroptosis. In vivo experiments provided compelling evidence supporting the potential of PAL as a potent therapeutic strategy for OS treatment. Conclusion: High-dose PAL-induced sustained oxidative stress by simultaneously targeting NRF2 inactivation and GPX4 degradation, establishing redox imbalance as a critical ferroptotic checkpoint in OS therapy. Antioxid. Redox Signal. 00, 000-000.

目的:等离子体活化液(PAL)是冷-常压等离子体(CAP)的一种间接应用形式,是一种产生活性氧和活性氮的电离气体,已被提出作为一种创新的治疗多种癌症的方法。尽管越来越多的证据表明PAL通过多种机制诱导细胞死亡,但铁下沉(一种由铁和脂质过氧化驱动的细胞死亡形式)在骨肉瘤(OS)中的作用仍然未知。结果:用CAP对液体进行不同时间的活化,产生不同剂量的PAL, PAL的抗肿瘤效果与剂量和治疗时间直接相关,并通过提高细胞内活性氧水平来实现。通过筛选三种有效剂量的PAL,我们发现PAL显著影响OS细胞的迁移和侵袭能力。蛋白质组学测序显示,pal处理组中几种与铁中毒相关的抗氧化蛋白增加。随后的研究结果表明,PAL调节核因子红系2相关因子2 (NRF2)及其下游的铁凋亡相关基因,主要通过消耗谷胱甘肽过氧化物酶4 (GPX4)在人OS细胞中诱导铁凋亡。最后,利用OS异种移植模型,我们发现PAL通过铁下垂在体内有效抑制肿瘤生长。创新:我们的研究强调了NRF2/GPX4轴作为pal诱导的铁下垂的关键途径的重要性。体内实验提供了令人信服的证据,支持PAL作为OS治疗的有效治疗策略的潜力。结论:大剂量pal通过同时靶向NRF2失活和GPX4降解诱导持续氧化应激,建立氧化还原失衡作为OS治疗中关键的铁稳定性检查点。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Dose-Dependent Ferroptosis Induction in Osteosarcoma via NRF2/GPX4 Axis Modulation by Plasma-Activated Liquid Therapy.","authors":"Longze Xiao, Mian Zhang, Peng He, Hao Wang, Sheng Li, Yonghong Wu, Shenglin Xu, Yong Hu","doi":"10.1177/15230864251399168","DOIUrl":"https://doi.org/10.1177/15230864251399168","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Plasma-activated liquid (PAL), an indirect application form of cold-atmospheric plasma (CAP)-an ionized gas generating reactive oxygen and nitrogen species, has been proposed as an innovative therapeutic approach for various cancer types. Despite accumulating evidence suggesting that PAL induces cell death through multiple mechanisms, the involvement of ferroptosis, a form of cell death driven by iron and lipid peroxidation, in osteosarcoma (OS) remains predominantly unknown. <b><i>Results:</i></b> CAP was used to activate the liquid for various durations, resulting in different doses of PAL. The antitumor efficacy of PAL was directly correlated with both the dosage and duration of treatment and was achieved by increasing the level of intracellular reactive oxygen species. Through screening three effective PAL doses, we discovered that PAL significantly influenced the migration and invasion capabilities of OS cells. Proteomic sequencing revealed increases in several ferroptosis-related antioxidant proteins in the PAL-treated group. Subsequent findings revealed that PAL modulated nuclear factor erythroid 2-related factor 2 (NRF2) and its downstream ferroptosis-related genes, predominantly resulting in the induction of ferroptosis by depleting glutathione peroxidase 4 (GPX4) in human OS cells. Finally, utilizing an OS xenograft model, we found that PAL effectively suppressed tumor growth <i>in vivo via</i> ferroptosis. <b><i>Innovation:</i></b> Our study highlights the importance of the NRF2/GPX4 axis as a pivotal pathway in PAL-induced ferroptosis. <i>In vivo</i> experiments provided compelling evidence supporting the potential of PAL as a potent therapeutic strategy for OS treatment. <b><i>Conclusion:</i></b> High-dose PAL-induced sustained oxidative stress by simultaneously targeting NRF2 inactivation and GPX4 degradation, establishing redox imbalance as a critical ferroptotic checkpoint in OS therapy. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145628097","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
PGK1 Persulfidation Promotes the Proliferation and Metastasis of Breast Cancer. PGK1过硫化促进乳腺癌的增殖和转移。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-12 DOI: 10.1177/15230864251394334
Chenghua Luo, Mengmeng Zhao, Yalu Wang, Yuxiang Xu, Shuai Chen, Weihua Liang, Kaige Yang, Jianming Hu

Aims: Endogenous hydrogen sulfide (H2S) is involved in the occurrence and development of breast cancer, while its underlying mechanism is not yet clear. Here, we aimed to focus on the molecular mechanism of endogenous H2S promoting the proliferation and metastasis of breast cancer. Results: In this study, four major findings were revealed: (1) Inhibition of cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE) increased the content of glucose in the supernatant of breast cancer cell and decreased the production of intracellular lactic acid and adenosine triphosphate. (2) Phosphoglycerate kinase 1 (PGK1) was persulfidated at Cys108 and Cys316, and its persulfidation level in breast cancer tissue was significantly higher than that in paracancerous tissue. (3) Blocking the persulfidation of PGK1 inhibited glycolysis and malignant biological behaviors of breast cancer cell. (4) The CSE inhibitor reduced the persulfidation of PGK1 and inhibited the growth and metastasis of xenograft tumors, whereas sodium hydrosulfide reversed the effect of CSE inhibitor. Preface PGK1 is not the only potential target for persulfidation. Innovation and Conclusion: This study revealed a novel mechanism involved in the upregulation of endogenous H2S in breast cancer. Endogenous H2S regulates glycolysis of breast cancer cells by mediating PGK1 persulfidation modification at Cys108 and Cys316, thereby promoting tumor proliferation and metastasis. This study offers a potential therapeutic strategy through targeting the upregulated endogenous H2S and persulfidation of PGK1. Antioxid. Redox Signal. 00, 000-000.

目的:内源性硫化氢(H2S)参与乳腺癌的发生发展,但其潜在机制尚不清楚。本研究旨在探讨内源性H2S促进乳腺癌增殖转移的分子机制。结果:(1)抑制胱硫氨酸-β-合成酶(CBS)和胱硫氨酸-γ-裂解酶(CSE)可提高乳腺癌细胞上清中葡萄糖的含量,降低细胞内乳酸和三磷酸腺苷的生成。(2)磷酸甘油酸激酶1 (PGK1)在Cys108和Cys316位点被过硫化,且其在乳腺癌组织中的过硫化水平显著高于癌旁组织。(3)阻断PGK1过硫化可抑制乳腺癌细胞糖酵解和恶性生物学行为。(4) CSE抑制剂降低PGK1过硫化,抑制异种移植物肿瘤的生长和转移,而氢硫化钠逆转了CSE抑制剂的作用。PGK1并不是过硫化的唯一潜在靶点。创新与结论:本研究揭示了乳腺癌内源性H2S上调的新机制。内源性H2S通过介导PGK1在Cys108和Cys316位点的过硫化修饰,调控乳腺癌细胞糖酵解,从而促进肿瘤的增殖和转移。该研究通过靶向上调的内源性H2S和PGK1过硫化提供了一种潜在的治疗策略。Antioxid。氧化还原信号:00000 - 00000。
{"title":"PGK1 Persulfidation Promotes the Proliferation and Metastasis of Breast Cancer.","authors":"Chenghua Luo, Mengmeng Zhao, Yalu Wang, Yuxiang Xu, Shuai Chen, Weihua Liang, Kaige Yang, Jianming Hu","doi":"10.1177/15230864251394334","DOIUrl":"https://doi.org/10.1177/15230864251394334","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Endogenous hydrogen sulfide (H<sub>2</sub>S) is involved in the occurrence and development of breast cancer, while its underlying mechanism is not yet clear. Here, we aimed to focus on the molecular mechanism of endogenous H<sub>2</sub>S promoting the proliferation and metastasis of breast cancer. <b><i>Results:</i></b> In this study, four major findings were revealed: (1) Inhibition of cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE) increased the content of glucose in the supernatant of breast cancer cell and decreased the production of intracellular lactic acid and adenosine triphosphate. (2) Phosphoglycerate kinase 1 (PGK1) was persulfidated at Cys108 and Cys316, and its persulfidation level in breast cancer tissue was significantly higher than that in paracancerous tissue. (3) Blocking the persulfidation of PGK1 inhibited glycolysis and malignant biological behaviors of breast cancer cell. (4) The CSE inhibitor reduced the persulfidation of PGK1 and inhibited the growth and metastasis of xenograft tumors, whereas sodium hydrosulfide reversed the effect of CSE inhibitor. Preface PGK1 is not the only potential target for persulfidation. <b><i>Innovation and Conclusion:</i></b> This study revealed a novel mechanism involved in the upregulation of endogenous H<sub>2</sub>S in breast cancer. Endogenous H<sub>2</sub>S regulates glycolysis of breast cancer cells by mediating PGK1 persulfidation modification at Cys108 and Cys316, thereby promoting tumor proliferation and metastasis. This study offers a potential therapeutic strategy through targeting the upregulated endogenous H<sub>2</sub>S and persulfidation of PGK1. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"0"},"PeriodicalIF":6.1,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145538862","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
Non-thermal Plasma-Derived Reactive Oxygen Species Induces Cell Death in Radioresistant Head and Neck Cancer via the c-MET/STAT3 Pathway. 非热等离子体源性活性氧通过c-MET/STAT3途径诱导放射耐药头颈癌细胞死亡
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-05 DOI: 10.1177/15230864251393938
Chan Oh, Mi Ae Lim, Shengzhe Cui, Yudan Piao, Sicong Zheng, Yan Li Jin, Shan Shen, Quoc Khanh Nguyen, Se-Hee Park, Young Il Kim, Jae Won Chang, Ho-Ryun Won, Ji Won Kim, Seung-Nam Jung, Bon Seok Koo

Aims: Radiation therapy is a crucial treatment modality for head and neck squamous cell carcinomas (HNSCCs). However, acquired radiation resistance due to various mechanisms poses a major clinical challenge for therapeutic strategies. Intriguingly, reactive oxygen species (ROS) are versatile signaling molecules that promote various cellular functions at low concentrations but induce cell death at above-critical threshold levels. Results: Here, we found that radioresistant (RR) cancer cells exhibited reduced ROS levels and activation of the mesenchymal-epithelial transition factor/signal transducer and activator of transcription 3 (c-MET/STAT3) pathway. To target common vulnerabilities in RR cancers, we applied ROS enhancement therapy using nonthermal plasma-activated media (NTPAM), a novel approach that effectively inhibits the viability of RR cancer cells and is associated with inactivation of the c-MET/STAT3 pathway. Mechanistically, the downregulation of total c-MET is related to ROS-mediated lysosomal degradation. In addition, NTPAM suppressed tumor growth in a mouse model of RR cancer, concurrently reducing the levels of both the total and activated forms of c-MET and decreasing STAT3 phosphorylation. Innovations and Conclusions: These findings suggest that ROS enhancement therapy can overcome radiation resistance, thereby offering a compelling rationale for considering NTPAM as a stand-alone or complementary therapeutic approach for treating patients with HNSCCs. Antioxid. Redox Signal. 00, 000-000.

目的:放射治疗是头颈部鳞状细胞癌(HNSCCs)的重要治疗方式。然而,由于各种机制引起的获得性放射耐药对治疗策略提出了重大的临床挑战。有趣的是,活性氧(ROS)是一种多用途的信号分子,在低浓度下促进各种细胞功能,但在高于临界阈值水平时诱导细胞死亡。结果:研究人员发现,放射耐药(RR)癌细胞表现出ROS水平降低和间充质-上皮过渡因子/信号换能器和转录激活因子3 (c-MET/STAT3)通路的激活。为了针对RR癌症的常见弱点,我们使用非热等离子体激活介质(NTPAM)应用ROS增强治疗,这是一种有效抑制RR癌细胞活力的新方法,并与c-MET/STAT3途径失活相关。机制上,总c-MET的下调与ros介导的溶酶体降解有关。此外,NTPAM抑制了RR癌小鼠模型中的肿瘤生长,同时降低了c-MET总形式和激活形式的水平,并降低了STAT3磷酸化。创新和结论:这些发现表明ROS增强疗法可以克服放射耐药,从而为考虑NTPAM作为治疗HNSCCs患者的独立或补充治疗方法提供了令人信服的理由。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Non-thermal Plasma-Derived Reactive Oxygen Species Induces Cell Death in Radioresistant Head and Neck Cancer <i>via</i> the c-MET/STAT3 Pathway.","authors":"Chan Oh, Mi Ae Lim, Shengzhe Cui, Yudan Piao, Sicong Zheng, Yan Li Jin, Shan Shen, Quoc Khanh Nguyen, Se-Hee Park, Young Il Kim, Jae Won Chang, Ho-Ryun Won, Ji Won Kim, Seung-Nam Jung, Bon Seok Koo","doi":"10.1177/15230864251393938","DOIUrl":"https://doi.org/10.1177/15230864251393938","url":null,"abstract":"<p><p><b><i>Aims</i></b>: Radiation therapy is a crucial treatment modality for head and neck squamous cell carcinomas (HNSCCs). However, acquired radiation resistance due to various mechanisms poses a major clinical challenge for therapeutic strategies. Intriguingly, reactive oxygen species (ROS) are versatile signaling molecules that promote various cellular functions at low concentrations but induce cell death at above-critical threshold levels. <b><i>Results:</i></b> Here, we found that radioresistant (RR) cancer cells exhibited reduced ROS levels and activation of the mesenchymal-epithelial transition factor/signal transducer and activator of transcription 3 (c-MET/STAT3) pathway. To target common vulnerabilities in RR cancers, we applied ROS enhancement therapy using nonthermal plasma-activated media (NTPAM), a novel approach that effectively inhibits the viability of RR cancer cells and is associated with inactivation of the c-MET/STAT3 pathway. Mechanistically, the downregulation of total c-MET is related to ROS-mediated lysosomal degradation. In addition, NTPAM suppressed tumor growth in a mouse model of RR cancer, concurrently reducing the levels of both the total and activated forms of c-MET and decreasing STAT3 phosphorylation. <b><i>Innovations and Conclusions:</i></b> These findings suggest that ROS enhancement therapy can overcome radiation resistance, thereby offering a compelling rationale for considering NTPAM as a stand-alone or complementary therapeutic approach for treating patients with HNSCCs. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145450364","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