Antioxidants & redox signaling最新文献

筛选
英文 中文
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 5.9 2区 生物学
Antioxidants & redox signaling Pub 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
{"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":"https://doi.org/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> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":5.9,"publicationDate":"2025-07-09","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
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 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-08 DOI: 10.1089/ars.2024.0868
Ruixue Sang, Xia Zhao, Ketao Sun, Yan Zhang, Bing Luo
{"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":"https://doi.org/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> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":5.9,"publicationDate":"2025-07-08","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
Thioredoxin-1 Downregulation in the SNpc Exacerbates the Cognitive Impairment Induced by MPTP. SNpc中硫氧还蛋白-1的下调加剧了MPTP诱导的认知障碍。
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-03-26 DOI: 10.1089/ars.2024.0630
Xianwen Zhang, Fang Yan, Xiong Jie He, Yali Chen, Rou Gu, Xianghuan Dong, Yonghang Wei, Liping Bai, Jie Bai
{"title":"Thioredoxin-1 Downregulation in the SNpc Exacerbates the Cognitive Impairment Induced by MPTP.","authors":"Xianwen Zhang, Fang Yan, Xiong Jie He, Yali Chen, Rou Gu, Xianghuan Dong, Yonghang Wei, Liping Bai, Jie Bai","doi":"10.1089/ars.2024.0630","DOIUrl":"10.1089/ars.2024.0630","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Parkinson's disease (PD) is characterized by dopaminergic (DAergic) neuron degeneration in the substantia nigra pars compacta (SNpc). Thioredoxin-1 (Trx-1) is a redox protein that protects neurons from various injuries. Our study revealed that Trx-1 overexpression improved the learning and memory impairments induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). However, the role of the specific transmission of signals from the SNpc to the hippocampus regulated by Trx-1 in cognition deficits associated with PD is still unknown. <b><i>Results:</i></b> We observed that Trx-1 downregulation in the SNpc aggravated cognitive dysfunction induced by MPTP. Importantly, we observed that the SNpc directly projects to the hippocampus. We found that the loss of DAergic neurons in the SNpc induced by MPTP resulted in a decrease in dopamine D1 receptor (D1R) expression in the hippocampus, which was promoted by Trx-1 downregulation in the SNpc. The levels of phosphorylated extracellular signal-regulated kinase (p-ERK1/2), phosphorylated cAMP-response element binding protein (p-CREB), brain-derived neurotrophic factor (BDNF), and postsynaptic density protein 95 (PSD95) in the hippocampus were decreased by MPTP and further decreased by Trx-1 downregulation in the SNpc. Finally, the number of synapses in the hippocampus was decreased by MPTP in the hippocampus and further reduced by Trx-1 downregulation in the SNpc. <b><i>Innovation:</i></b> Trx-1 downregulation accelerated the loss of DAergic neurons in the SNpc, leading to a decrease in the number dopaminergic projections to the hippocampus, subsequently inhibiting the D1R-ERK1/2-CREB-BDNF pathway in the hippocampus, and ultimately impairing hippocampus-dependent cognition. <b><i>Conclusions:</i></b> These results indicate that a decrease in Trx-1 level in the SNpc plays a critical regulatory role in cognitive dysfunction in individuals with PD by decreasing the hippocampal D1R signaling pathway. <i>Antioxid. Redox Signal.</i> 43, 138-150.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"138-150"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143707900","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
Impacts of Radiation on Metabolism and Vascular Cell Senescence. 辐射对代谢和血管细胞衰老的影响。
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-04-16 DOI: 10.1089/ars.2024.0741
Junichi Abe, Khanh Chau, Anahita Mojiri, Guangyu Wang, Masayoshi Oikawa, Venkata S K Samanthapudi, Abigail M Osborn, Keila C Ostos-Mendoza, Karla N Mariscal-Reyes, Tammay Mathur, Abhishek Jain, Joerg Herrmann, Syed Wamique Yusuf, Sunil Krishnan, Anita Deswal, Steven H Lin, Sivareddy Kotla, John P Cooke, Nhat-Tu Le
{"title":"Impacts of Radiation on Metabolism and Vascular Cell Senescence.","authors":"Junichi Abe, Khanh Chau, Anahita Mojiri, Guangyu Wang, Masayoshi Oikawa, Venkata S K Samanthapudi, Abigail M Osborn, Keila C Ostos-Mendoza, Karla N Mariscal-Reyes, Tammay Mathur, Abhishek Jain, Joerg Herrmann, Syed Wamique Yusuf, Sunil Krishnan, Anita Deswal, Steven H Lin, Sivareddy Kotla, John P Cooke, Nhat-Tu Le","doi":"10.1089/ars.2024.0741","DOIUrl":"10.1089/ars.2024.0741","url":null,"abstract":"<p><p><b><i>Significance:</i></b> This review investigates how radiation therapy (RT) increases the risk of delayed cardiovascular disease (CVD) in cancer survivors. Understanding the mechanisms underlying radiation-induced CVD is essential for developing targeted therapies to mitigate these effects and improve long-term outcomes for patients with cancer. <b><i>Recent Advances:</i></b> Recent studies have primarily focused on metabolic alterations induced by irradiation in various cancer cell types. However, there remains a significant knowledge gap regarding the role of chronic metabolic alterations in normal cells, particularly vascular cells, in the progression of CVD after RT. <b><i>Critical Issues:</i></b> This review centers on RT-induced metabolic alterations in vascular cells and their contribution to senescence accumulation and chronic inflammation across the vasculature post-RT. We discuss key metabolic pathways, including glycolysis, the tricarboxylic acid cycle, lipid metabolism, glutamine metabolism, and redox metabolism (nicotinamide adenine dinucleotide/Nicotinamide adenine dinucleotide (NADH) and nicotinamide adenine dinucleotide phosphate (NADP<sup>+</sup>)/NADPH). We further explore the roles of regulatory proteins such as p53, adenosine monophosphate-activated protein kinase, and mammalian target of rapamycin in driving these metabolic dysregulations. The review emphasizes the impact of immune-vascular crosstalk mediated by the senescence-associated secretory phenotype, which perpetuates metabolic dysfunction, enhances chronic inflammation, drives senescence accumulation, and causes vascular damage, ultimately contributing to cardiovascular pathogenesis. <b><i>Future Directions:</i></b> Future research should prioritize identifying therapeutic targets within these metabolic pathways or the immune-vascular interactions influenced by RT. Correcting metabolic dysfunction and reducing chronic inflammation through targeted therapies could significantly improve cardiovascular outcomes in cancer survivors. <i>Antioxid. Redox Signal.</i> 43, 92-114.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"92-114"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144061398","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 Mediates the Progression of Hyperuricemic Nephropathy by Activating RAGE Signaling. 铁下垂通过激活RAGE信号介导高尿酸血症肾病的进展。
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-03-10 DOI: 10.1089/ars.2024.0672
Qiang Wang, Yuemei Xi, Hairong Zhao, De Xie, Linqian Yu, Yunbo Yan, Jiayu Chen, Qian Zhang, Meng Liang, Jidong Cheng
{"title":"Ferroptosis Mediates the Progression of Hyperuricemic Nephropathy by Activating RAGE Signaling.","authors":"Qiang Wang, Yuemei Xi, Hairong Zhao, De Xie, Linqian Yu, Yunbo Yan, Jiayu Chen, Qian Zhang, Meng Liang, Jidong Cheng","doi":"10.1089/ars.2024.0672","DOIUrl":"10.1089/ars.2024.0672","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Hyperuricemic nephropathy (HN) represents a prevalent complication of hyperuricemia, typified by tubular dysfunction, inflammation, and progressive renal fibrosis with unclear mechanisms. Ferroptosis, an iron-dependent regulated cell death, is implicated in multiple diseases, but has rarely been linked to HN. In this study, we aim to explore the possible role of ferroptosis in HN and its underlying mechanisms. <b><i>Results:</i></b> We showed that urate oxidase knockout mice, a model of hyperuricemia, exhibited renal impairment with elevated uric acid, creatinine, and blood urea nitrogen levels, accompanied by increased iron deposition and decreased glutathione peroxidase 4 (GPX4) and xCT expressions, suggesting ferroptosis involvement. Ferroptosis inhibitor Ferrostatin-1 (Fer-1) ameliorated renal injury, inflammatory cell infiltration, and fibrosis in these mice. Mechanistically, Fer-1 restored antioxidant protein levels, normalized ferroptosis-associated protein expressions, diminished iron overload and lipid peroxidation, and suppressed inflammatory markers and mitogen-activated protein kinase signaling. <i>In vitro</i>, monosodium urate crystals induced ferroptosis in human kidney 2 cells, characterized by increased lipid peroxidation and iron accumulation. Notably, receptor for advanced glycation end products (RAGE) inhibition alleviated renal injury, inflammation, and fibrosis albeit without directly diminishing ferroptosis. These findings were validated in human hyperuricemia-related kidney disease samples showing increased iron deposition, decreased GPX4, and elevated RAGE expression. <b><i>Innovation and Conclusion:</i></b> This study suggests that ferroptosis may play a role in the development of renal injury, inflammation, and fibrosis in HN, potentially mediated through RAGE signaling. While RAGE inhibition improved renal injury, it did not directly affect ferroptosis, indicating a complex and context-dependent role of RAGE in kidney injury. These findings highlight ferroptosis and its associated pathways, including RAGE signaling, as potential therapeutic targets for HN. <i>Antioxid. Redox Signal.</i> 43, 56-74.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"56-74"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143584354","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
The Dual Role of Active Site Hydroxylated Residue in Peroxiredoxin Sulfinylation Catalysis. 活性位点羟基化残基在过氧还氧素亚砜化催化中的双重作用。
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-03-07 DOI: 10.1089/ars.2024.0685
Julie Mathieu, Alexandre Kriznik, Christophe Charron, Romain Perchat-Varlet, Benjamin Selles, Sophie Rahuel-Clermont
{"title":"The Dual Role of Active Site Hydroxylated Residue in Peroxiredoxin Sulfinylation Catalysis.","authors":"Julie Mathieu, Alexandre Kriznik, Christophe Charron, Romain Perchat-Varlet, Benjamin Selles, Sophie Rahuel-Clermont","doi":"10.1089/ars.2024.0685","DOIUrl":"10.1089/ars.2024.0685","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Peroxiredoxins (Prx) are ubiquitous Cys peroxidases regulated by sulfinylation, a modification that occurs when the sulfenic acid generated on the catalytic Cys by peroxide reduction reacts with a second molecule of peroxide. In the Prx1 family, sulfinylation sensitivity is controlled by competition between a structural transition from a fully folded (FF) to locally unfolded (LU) conformation and the chemical step of sulfinylation. The initial peroxide reduction relies on a conserved catalytic hydroxylated residue that allows peroxide optimal activation. This study aimed at investigating the role of this catalytic residue in sulfinylation. <b><i>Results:</i></b> Sulfenate attack on peroxide was favored by one order of magnitude when a catalytic Thr was present, for yeast cytosolic Prx1-type enzymes, human Prx1 and yeast mitochondrial Prx, a Prx6-type enzyme. Furthermore, pKa determination supported the notion of electrostatic interaction between the catalytic hydroxyl and sulfenate intermediate. Finally, FF-LU transition kinetics was faster with a catalytic Thr, supporting that the hydroxyl group proximity to the nascent sulfenate group also promotes the FF-LU transition. <b><i>Innovation:</i></b> We identify a major mechanism that activates sulfinylation in hyperoxidation-sensitive Prxs from the Prx1 and Prx6 families. Furthermore, we show that the catalytic hydroxylated residue holds a dual role in regulating hyperoxidation sensitivity, by activating the sulfinylation reaction, while also promoting the competing FF to LU transition, thus acting as an important regulatory determinant. <b><i>Conclusion:</i></b> The present work sets the basis for investigating other instances of Cys proteins regulated by sulfinylation, a modification increasingly recognized in cell redox regulation and signaling. <i>Antioxid. Redox Signal.</i> 43, 1-13.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"1-13"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143595837","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
GYY4137, a Slow-Releasing Hydrogen Sulfide Donor, Attenuates Skeletal Muscle Abnormalities in a Murine Model of Duchenne Muscular Dystrophy. GYY4137,一种缓释硫化氢供体,在杜氏肌营养不良小鼠模型中减轻骨骼肌异常。
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-06-05 DOI: 10.1089/ars.2024.0702
Małgorzata Myszka, Ewa Jakubczak, Olga Mucha, Kalina Hajok, Urszula Waśniowska, Anna Nalepa, Józef Dulak, Agnieszka Łoboda
{"title":"GYY4137, a Slow-Releasing Hydrogen Sulfide Donor, Attenuates Skeletal Muscle Abnormalities in a Murine Model of Duchenne Muscular Dystrophy.","authors":"Małgorzata Myszka, Ewa Jakubczak, Olga Mucha, Kalina Hajok, Urszula Waśniowska, Anna Nalepa, Józef Dulak, Agnieszka Łoboda","doi":"10.1089/ars.2024.0702","DOIUrl":"10.1089/ars.2024.0702","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Duchenne muscular dystrophy (DMD) is a severe, incurable X-linked genetic disorder caused by mutations in the <i>DMD</i> gene, leading to a deficiency of the muscle structural protein, dystrophin, which results in damage to skeletal and cardiac muscles. Altered expression of enzymes that generate hydrogen sulfide (H<sub>2</sub>S) has been demonstrated in dystrophic muscles, however, the exact role of this gasotransmitter in DMD remains elusive. Here, we investigated the effect of the slow-releasing H<sub>2</sub>S donor (GYY4137) on the skeletal muscles of the dystrophin-deficient <i>mdx</i> mice. <b><i>Methods and Results:</i></b> Grip strength assay and the treadmill exhaustion test showed that administering the GYY4137 donor to <i>mdx</i> mice improved DMD-related decline in motor functions. Additionally, the H<sub>2</sub>S donor decreased the level of muscle damage markers such as lactate dehydrogenase, creatine kinase, and osteopontin (OPN). Histological, gene, and protein analyses of the dystrophic gastrocnemius and diaphragm muscles revealed reduced inflammation and fibrosis after treatment with the H<sub>2</sub>S donor. Moreover, we showed decreased necrosis with improved muscle regeneration and angiogenesis. We demonstrated that GYY4137 upregulates the levels of phosphorylated AMPKα, as well as the cytoprotective and antioxidant heme oxygenase-1, mitochondrial superoxide dismutase, and glutamate-cysteine ligase modifier subunit (<i>Gclm</i>). Finally, it exerted an anti-apoptotic effect by reducing cleaved caspase-3 and caspase-3 and increasing AKT phosphorylation. <b><i>Innovation and Conclusion:</i></b> The administration of GYY4137 improves exercise capacity and ameliorates the markers of inflammation, fibrosis, oxidative stress, apoptosis, and necrosis in the skeletal muscles of <i>mdx</i> animals pointing out its possible therapeutic use in DMD pathology. <i>Antioxid. Redox Signal.</i> 43, 115-137.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"115-137"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144232955","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
ALOX15 Aggravates Metabolic Dysfunction-Associated Steatotic Liver Disease in Mice with Type 2 Diabetes via Activating the PPARγ/CD36 Axis. ALOX15通过激活PPARγ/CD36轴加重2型糖尿病小鼠代谢功能障碍相关的脂肪变性肝病
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-01-16 DOI: 10.1089/ars.2024.0670
Wenhui Yan, Xin Cui, Tingli Guo, Na Liu, Zhuanzhuan Wang, Yuzhuo Sun, Yuanrui Shang, Jieyun Liu, Yuanyuan Zhu, Yangyang Zhang, Lina Chen
{"title":"ALOX15 Aggravates Metabolic Dysfunction-Associated Steatotic Liver Disease in Mice with Type 2 Diabetes via Activating the PPARγ/CD36 Axis.","authors":"Wenhui Yan, Xin Cui, Tingli Guo, Na Liu, Zhuanzhuan Wang, Yuzhuo Sun, Yuanrui Shang, Jieyun Liu, Yuanyuan Zhu, Yangyang Zhang, Lina Chen","doi":"10.1089/ars.2024.0670","DOIUrl":"10.1089/ars.2024.0670","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent hepatic disorder worldwide. Arachidonic acid 15-lipoxygenase (ALOX15), an enzyme catalyzing the peroxidation of polyunsaturated fatty acids, plays a crucial role in various diseases. Here, we sought to investigate the involvement of ALOX15 in MASLD. <b><i>Results:</i></b> In this study, we observed upregulation of ALOX15 in the liver of high-fat diet (HFD)- and streptozotocin (STZ)-induced mice. Metabolomic analysis revealed elevated levels of ALOX15 metabolites, 12(S)-hydroperoxyeicosatetraenoic acid and 15(S)-hydroperoxyeicosatetraenoic acid. Transcriptomic analysis showed that the increased fatty acid uptake regulated by the PPARγ/CD36 pathway predominated in lipid accumulation. To elucidate the mechanism underlying ALOX15-induced lipid accumulation, HepG2 cells were transfected with a lentivirus expressing ALOX15 or small interfering RNA targeting ALOX15 and exposed to palmitic acid (PA). Both ALOX15 overexpression and PA exposure led to increased intracellular free fatty acid and triglyceride, resulting in lipotoxicity. ALOX15 overexpression aggravated the effect of PA, while the knockdown of ALOX15 attenuated PA-induced lipotoxicity. Moreover, the treatment with PPARγ antagonist GW9662 or CD36 inhibitor sulfosuccinimidyl oleate sodium effectively reduced lipid accumulation and lipotoxicity resulting from ALOX15 overexpression and PA exposure, indicating the involvement of the PPARγ/CD36 pathway in ALOX15-mediated lipid accumulation. Furthermore, liraglutide, a widely used glucagon-like peptide 1 receptor (GLP-1R) agonist (GLP-1RA), improved hepatic lipid accumulation in HFD/STZ-induced mice by suppressing the ALOX15/PPARγ/CD36 pathway. <b><i>Innovation and Conclusion:</i></b> Our study underscores the potential of ALOX15 as an emerging therapeutic target for MASLD. In addition, the GLP-1RA may confer hepatoprotection by regulating ALOX15, enhancing our comprehension of the mechanisms underpinning their protection on MASLD. <i>Antioxid. Redox Signal.</i> 43, 37-55.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"37-55"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998899","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
Functions of Endogenously Produced and Exogenously Applied Melatonin in Higher Plants. 高等植物内源和外源褪黑素的功能。
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-06-11 DOI: 10.1089/ars.2024.0889
Francisco J Corpas, Jorge Taboada, Rosa M Rivero, Russel J Reiter, José M Palma
{"title":"Functions of Endogenously Produced and Exogenously Applied Melatonin in Higher Plants.","authors":"Francisco J Corpas, Jorge Taboada, Rosa M Rivero, Russel J Reiter, José M Palma","doi":"10.1089/ars.2024.0889","DOIUrl":"10.1089/ars.2024.0889","url":null,"abstract":"<p><p><b><i>Significance:</i></b> The role of melatonin (MEL) in plants has gained significant relevance due to its involvement in a wide range of physiological functions, particularly in response mechanisms to both abiotic and biotic stresses. <b><i>Recent Advances:</i></b> Recent progress highlights the significance of the biosynthetic pathway of MEL in plants, which surpasses that of animals. The discovery of specific plant MEL receptors has revealed new signaling mechanisms. Studies also show that applying exogenous MEL offers benefits under stress conditions and helps maintain the organoleptic qualities of fruits and vegetables during postharvest storage. <b><i>Critical Issues:</i></b> This review explores MEL's biochemistry, emphasizing its dual role as both an antioxidant and a signaling molecule. It examines how MEL interacts with phytohormones, its role in regulating the metabolism of reactive oxygen and nitrogen species, and its influence on plant growth and stress tolerance. The potential of MEL-based biotechnological applications for enhancing crop resilience and postharvest quality is also discussed. <b><i>Future Directions:</i></b> Future research should prioritize molecular mechanisms, high-throughput approaches, and translational studies to bridge the gap between fundamental science and agricultural practices. MEL's role as a sustainable solution in agriculture offers exciting possibilities for addressing global food security challenges. <i>Antioxid. Redox Signal.</i> 43, 151-188.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"151-188"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144265069","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
Bach1 Deficiency Ameliorates Radiation Pneumonitis via Activating TFAM Signaling Pathway. Bach1缺乏通过激活TFAM信号通路改善放射性肺炎
IF 5.9 2区 生物学
Antioxidants & redox signaling Pub Date : 2025-07-01 Epub Date: 2025-03-10 DOI: 10.1089/ars.2024.0742
Jianfeng Huang, Yanli Zhang, Fengjuan Jiang, Yaru Zhang, Shengpeng Li, Shuai He, Jiaojiao Sun, Dan Chen, Qingfeng Pang, Yaxian Wu
{"title":"Bach1 Deficiency Ameliorates Radiation Pneumonitis via Activating TFAM Signaling Pathway.","authors":"Jianfeng Huang, Yanli Zhang, Fengjuan Jiang, Yaru Zhang, Shengpeng Li, Shuai He, Jiaojiao Sun, Dan Chen, Qingfeng Pang, Yaxian Wu","doi":"10.1089/ars.2024.0742","DOIUrl":"10.1089/ars.2024.0742","url":null,"abstract":"<p><p><b><i>Aims:</i></b> BTB and CNC homology 1 (Bach1) is a transcription factor that mediates oxidative stress and inflammation and participates in the progression of diseases such as atherosclerosis, colitis, and acute lung injury. In this study, we aimed to explore the role of Bach1 in radiation pneumonitis (RP) and elucidate its underlying mechanism. <b><i>Results:</i></b> Bach1 expression was significantly elevated in the lung tissues of RP mice. Deletion of the Bach1 gene markedly ameliorated X-ray-induced RP by reducing inflammation and oxidative stress. <i>In vitro</i> experiments demonstrated that Bach1 deficiency mitigated radiation-induced oxidative damage and inflammation in bone marrow-derived macrophages. Conversely, Bach1 overexpression exacerbated oxidative stress and inflammation in radiation-treated macrophages. Mechanistically, using the JASPAR database, electromobility shift assays, and luciferase reporter assays, we revealed that Bach1 inhibited mRNA expression of mitochondrial transcription factor A (<i>TFAM</i>) by directly binding to its promoter region. <b><i>Innovation and Conclusion:</i></b> Our findings indicate that silencing of Bach1 protects against RP by upregulating the mRNA expression of <i>TFAM</i>, which, in turn, enhances mitochondrial function and reduces inflammation and oxidative stress. This study provides valuable insights into potential therapeutic strategies for patients with RP through Bach1 inhibition. <i>Antioxid. Redox Signal.</i> 43, 75-91.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"75-91"},"PeriodicalIF":5.9,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143584353","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信