首页 > 最新文献

Antioxidants & redox signaling最新文献

英文 中文
Targeting the HNRNPA2B1/HDGF/PTN Axis to Overcome Radioresistance in Non-Small Cell Lung Cancer. 靶向HNRNPA2B1/HDGF/PTN轴克服非小细胞肺癌的放射耐药
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-08-01 Epub Date: 2025-07-11 DOI: 10.1089/ars.2024.0808
Fushi Han, Shuzhen Chen, Kangwei Zhang, Kunming Zhang, Meng Wang, Peijun Wang

Aims: Radioresistance in non-small cell lung cancer (NSCLC) presents a major barrier to effective treatment. This study explores the molecular mechanisms underlying this resistance, focusing on the heterogeneous nuclear ribonucleoprotein A2B1/hepatoma-derived growth factor/pleiotrophin (HNRNPA2B1/HDGF/PTN) signaling pathway and its role in autophagy-dependent ferroptosis regulation. Our aim is to uncover how this pathway contributes to tumor cell survival under radiotherapy stress, thereby identifying potential therapeutic targets to overcome radioresistance. Results: We developed radiotherapy-resistant lung cancer cell lines and assessed their proliferation and migration capabilities through Cell Counting Kit-8 and Transwell assays, respectively. Single-cell RNA sequencing revealed significant differences in gene expression profiles between radioresistance and radiation-sensitive cells. Functional studies, including immunofluorescence, flow cytometry, and biochemical staining, confirmed that radioresistance was associated with enhanced autophagy and altered ferroptosis. Furthermore, HNRNPA2B1 knockdown reduced the expression of Ki67 and proliferating cell nuclear antigen, markers of proliferation, in a mouse tumor model. In addition, modulation of HNRNPA2B1 affected protein interactions and N6-methyladenosine RNA modifications, as demonstrated by reverse transcription-quantitative polymerase chain reaction, Western blot, and methylation RNA immunoprecipitation-quantitative PCR. Innovation: This study provides new insights into how the HNRNPA2B1/HDGF/PTN pathway promotes radioresistance by influencing autophagy-dependent ferroptosis. This mechanism represents a potential vulnerability that could be therapeutically targeted to improve radiotherapy efficacy in lung cancer. Conclusion: Our findings demonstrate that the HNRNPA2B1/HDGF/PTN signaling pathway plays a crucial role in sustaining radioresistant phenotypes by modulating autophagy and ferroptosis. Targeting this pathway may enhance the therapeutic response in NSCLC, offering a novel strategy to combat treatment resistance. Antioxid. Redox Signal. 43, 189-214.

目的:非小细胞肺癌(NSCLC)的放射耐药是有效治疗的主要障碍。本研究探讨了这种耐药性的分子机制,重点研究了异质核核糖核蛋白A2B1/肝癌衍生生长因子/多营养因子(HNRNPA2B1/HDGF/PTN)信号通路及其在自噬依赖性铁凋亡调节中的作用。我们的目的是揭示这一途径如何促进肿瘤细胞在放疗应激下的存活,从而确定克服放射耐药的潜在治疗靶点。结果:我们开发了放疗耐药肺癌细胞系,并分别通过细胞计数试剂盒-8和Transwell检测评估了它们的增殖和迁移能力。单细胞RNA测序显示,辐射抗性细胞和辐射敏感细胞之间的基因表达谱存在显著差异。功能研究,包括免疫荧光、流式细胞术和生化染色,证实放射耐药与自噬增强和铁下垂改变有关。此外,在小鼠肿瘤模型中,HNRNPA2B1敲低可降低Ki67和增殖细胞核抗原(增殖标志物)的表达。此外,通过逆转录-定量聚合酶链反应、Western blot和甲基化RNA免疫沉淀-定量PCR证实,HNRNPA2B1的调节影响蛋白相互作用和n6 -甲基腺苷RNA修饰。创新:本研究为HNRNPA2B1/HDGF/PTN通路如何通过影响自噬依赖性铁凋亡促进辐射耐药提供了新的见解。这一机制代表了一种潜在的脆弱性,可以作为治疗目标来提高肺癌放射治疗的疗效。结论:我们的研究结果表明,HNRNPA2B1/HDGF/PTN信号通路通过调节自噬和铁凋亡在维持放射抗性表型中起着至关重要的作用。靶向这一途径可能会增强非小细胞肺癌的治疗反应,提供一种对抗治疗耐药性的新策略。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Targeting the HNRNPA2B1/HDGF/PTN Axis to Overcome Radioresistance in Non-Small Cell Lung Cancer.","authors":"Fushi Han, Shuzhen Chen, Kangwei Zhang, Kunming Zhang, Meng Wang, Peijun Wang","doi":"10.1089/ars.2024.0808","DOIUrl":"10.1089/ars.2024.0808","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Radioresistance in non-small cell lung cancer (NSCLC) presents a major barrier to effective treatment. This study explores the molecular mechanisms underlying this resistance, focusing on the heterogeneous nuclear ribonucleoprotein A2B1/hepatoma-derived growth factor/pleiotrophin (HNRNPA2B1/HDGF/PTN) signaling pathway and its role in autophagy-dependent ferroptosis regulation. Our aim is to uncover how this pathway contributes to tumor cell survival under radiotherapy stress, thereby identifying potential therapeutic targets to overcome radioresistance. <b><i>Results:</i></b> We developed radiotherapy-resistant lung cancer cell lines and assessed their proliferation and migration capabilities through Cell Counting Kit-8 and Transwell assays, respectively. Single-cell RNA sequencing revealed significant differences in gene expression profiles between radioresistance and radiation-sensitive cells. Functional studies, including immunofluorescence, flow cytometry, and biochemical staining, confirmed that radioresistance was associated with enhanced autophagy and altered ferroptosis. Furthermore, HNRNPA2B1 knockdown reduced the expression of Ki67 and proliferating cell nuclear antigen, markers of proliferation, in a mouse tumor model. In addition, modulation of HNRNPA2B1 affected protein interactions and N6-methyladenosine RNA modifications, as demonstrated by reverse transcription-quantitative polymerase chain reaction, Western blot, and methylation RNA immunoprecipitation-quantitative PCR. <b><i>Innovation:</i></b> This study provides new insights into how the HNRNPA2B1/HDGF/PTN pathway promotes radioresistance by influencing autophagy-dependent ferroptosis. This mechanism represents a potential vulnerability that could be therapeutically targeted to improve radiotherapy efficacy in lung cancer. <b><i>Conclusion:</i></b> Our findings demonstrate that the HNRNPA2B1/HDGF/PTN signaling pathway plays a crucial role in sustaining radioresistant phenotypes by modulating autophagy and ferroptosis. Targeting this pathway may enhance the therapeutic response in NSCLC, offering a novel strategy to combat treatment resistance. <i>Antioxid. Redox Signal.</i> 43, 189-214.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"189-214"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144607218","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
Sulforaphane Targets Multiple Pathological Processes in Friedreich Ataxia Patient-Induced Pluripotent Stem Cell-Derived Sensory Neurons. 萝卜硫素靶向弗里德里希共济失调患者诱导的多能干细胞来源的感觉神经元的多种病理过程。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-08-01 Epub Date: 2025-05-23 DOI: 10.1089/ars.2024.0756
Wenyao Yang, Bruce Thompson, Sara Miellet, Marnie Maddock, Marek Napierala, Mirella Dottori, Faith Kwa

Aims: In Friedreich ataxia (FRDA), early motor discoordination stems from dysfunctional sensory neurons in the spinal cord driven by epigenetic dysregulation, frataxin (FXN) deficiency, oxidative stress, and inflammation. Omaveloxolone, a nuclear factor erythroid 2-related factor-2 (NRF2) inducer, is the only treatment available. In various chronic disease models, sulforaphane (SF) can target NRF2 and the above processes. This study compared the effects of SF with omaveloxolone and dimethyl fumarate (DMF) in sensory neurons generated from FRDA patient-induced pluripotent stem cells and their isogenic control. Results: The successful generation of the FRDA and isogenic control sensory neurons was confirmed by the positive expression of β-III TUBULIN, BRN3A, ISLET1, PERIPHERIN, and tropomyosin receptor kinase C. In comparison with the isogenic control, FRDA sensory neurons displayed an aberrant gene expression profile alike to that reported in patients. None of the drugs affected the viability of the isogenic control sensory neurons. SF treatment improved the viability of FRDA sensory neurons by up to 61% versus the untreated control. DMF treatment showed a modest 35% increase, while omaveloxolone lacked an effect. SF-treated FRDA sensory neurons demonstrated increased reduced glutathione/oxidized glutathione ratio and expression of FXN and redox markers, and a reduced expression of selected epigenetic enzymes and inflammatory cytokines, at the respective gene and protein levels. DMF and omaveloxolone treatments only modulated some of these biomarkers. Innovation: We revealed the therapeutic potential of SF and how it performs in comparison with omaveloxolone and DMF, in a physiologically and genetically relevant in vitro FRDA model. Conclusion: SF offers a multipronged approach to alleviating the different cellular events underlying FRDA. Antioxid. Redox Signal. 43, 308-327.

目的:在弗里德赖希共济失调(FRDA)中,早期运动失调源于表观遗传失调、frataxin (FXN)缺乏、氧化应激和炎症驱动的脊髓感觉神经元功能失调。奥马维洛酮是一种核因子-红细胞2相关因子-2 (NRF2)诱导剂,是唯一可用的治疗方法。在多种慢性疾病模型中,萝卜硫素(sulforaphane, SF)可靶向NRF2及上述过程。本研究比较了SF与奥马洛酮和富马酸二甲酯(DMF)对FRDA患者诱导的多能干细胞产生的感觉神经元的影响及其等基因对照。结果:β-III TUBULIN、BRN3A、ISLET1、PERIPHERIN和原肌球蛋白受体激酶c的阳性表达证实了FRDA和等基因对照感觉神经元的成功生成。与等基因对照相比,FRDA感觉神经元表现出与患者相似的异常基因表达谱。所有药物均未影响等基因控制感觉神经元的活力。与未治疗的对照组相比,SF治疗可使FRDA感觉神经元的活力提高61%。DMF治疗显示适度的35%的增加,而奥马洛酮缺乏效果。sf处理的FRDA感觉神经元显示还原性谷胱甘肽/氧化性谷胱甘肽比例增加,FXN和氧化还原标记物的表达增加,特定表观遗传酶和炎症细胞因子的表达减少,在各自的基因和蛋白质水平上。DMF和奥马洛酮治疗只能调节其中的一些生物标志物。创新:在生理和遗传相关的体外FRDA模型中,我们揭示了SF的治疗潜力,以及它与奥马洛酮和DMF相比的表现。结论:SF提供了一种多管齐下的方法来缓解FRDA背后的不同细胞事件。Antioxid。氧化还原信号:00000 - 00000。[图:见正文]。
{"title":"Sulforaphane Targets Multiple Pathological Processes in Friedreich Ataxia Patient-Induced Pluripotent Stem Cell-Derived Sensory Neurons.","authors":"Wenyao Yang, Bruce Thompson, Sara Miellet, Marnie Maddock, Marek Napierala, Mirella Dottori, Faith Kwa","doi":"10.1089/ars.2024.0756","DOIUrl":"10.1089/ars.2024.0756","url":null,"abstract":"<p><p><b><i>Aims:</i></b> In Friedreich ataxia (FRDA), early motor discoordination stems from dysfunctional sensory neurons in the spinal cord driven by epigenetic dysregulation, frataxin (FXN) deficiency, oxidative stress, and inflammation. Omaveloxolone, a nuclear factor erythroid 2-related factor-2 (NRF2) inducer, is the only treatment available. In various chronic disease models, sulforaphane (SF) can target NRF2 and the above processes. This study compared the effects of SF with omaveloxolone and dimethyl fumarate (DMF) in sensory neurons generated from FRDA patient-induced pluripotent stem cells and their isogenic control. <b><i>Results:</i></b> The successful generation of the FRDA and isogenic control sensory neurons was confirmed by the positive expression of β-III TUBULIN, BRN3A, ISLET1, PERIPHERIN, and tropomyosin receptor kinase C. In comparison with the isogenic control, FRDA sensory neurons displayed an aberrant gene expression profile alike to that reported in patients. None of the drugs affected the viability of the isogenic control sensory neurons. SF treatment improved the viability of FRDA sensory neurons by up to 61% versus the untreated control. DMF treatment showed a modest 35% increase, while omaveloxolone lacked an effect. SF-treated FRDA sensory neurons demonstrated increased reduced glutathione/oxidized glutathione ratio and expression of FXN and redox markers, and a reduced expression of selected epigenetic enzymes and inflammatory cytokines, at the respective gene and protein levels. DMF and omaveloxolone treatments only modulated some of these biomarkers. <b><i>Innovation:</i></b> We revealed the therapeutic potential of SF and how it performs in comparison with omaveloxolone and DMF, in a physiologically and genetically relevant <i>in vitro</i> FRDA model. <b><i>Conclusion:</i></b> SF offers a multipronged approach to alleviating the different cellular events underlying FRDA. <i>Antioxid. Redox Signal.</i> 43, 308-327.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"308-327"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144126182","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
Redox Differences Between Neurons and Astrocytes In Vivo in Ischemic Brain Tissues of Rodents. 啮齿动物缺血脑组织中神经元和星形胶质细胞氧化还原差异
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-08-01 Epub Date: 2025-05-19 DOI: 10.1089/ars.2024.0876
Daria A Kotova, Aleksandra D Ivanova, Ilya V Kelmanson, Kseniia I Morozova, Yulia V Khramova, Maxim A Solotenkov, Evgeny A Stepanov, Aleksandr A Moshchenko, Alisa B Tiaglik, Anna A Fedotova, Anton V Zalygin, Vladimir A Oleinikov, Alexey G Katrukha, Alexey Semyanov, Vsevolod V Belousov, Andrei B Fedotov, Ilya V Fedotov, Nadezda A Brazhe, Dmitry S Bilan

Aims: Reactive oxygen species (ROS) are considered to play a key damaging role in brain during the development of ischemic stroke. To clarify how different ROS contribute to ischemic pathogenesis, innovative approaches for real-time in vivo detection of redox parameters are necessary. Results: Using highly sensitive genetically encoded biosensor HyPer7 and a fiber-optic neurointerface technology, we demonstrated that the level of hydrogen peroxide (H2O2) slowly increases in neurons and astrocytes of the ischemic area of the rat brain after middle cerebral artery occlusion during next 40 h; notably, in astrocytes the level is somewhat higher. Raman microspectroscopy in awake mice also revealed redox differences between mitochondria of neurons and astrocytes during acute ischemia caused by photothrombosis. Astrocytes demonstrated the overloading of the electron transport chain (ETC) with electrons after 1 h of ischemia, whereas neurons do not demonstrate changes in the amount of reduced electron carries. Innovation and Conclusion: The combination of novel in vivo approaches allows to detail redox events with spatiotemporal resolution. We demonstrated redox difference between neurons and astrocytes in damaged brain areas in vivo. An elevated loading of astrocytic ETC with electrons during the acute ischemia phase provides basis for the increased generation of superoxide anion radical (O2•-) with its following conversion to other reactive species. However, we observed increased H2O2 concentrations in astrocytes and neurons at later pathogenesis stages. During this period, ETC did not demonstrate an elevated loading with electrons, and therefore, increased H2O2 generation may be a phenomenon of secondary redox events. Antioxid. Redox Signal. 43, 272-287.

目的:活性氧(Reactive oxygen species, ROS)被认为在缺血性脑卒中的发展过程中起着关键的脑损伤作用。为了阐明不同的活性氧如何促进缺血发病机制,有必要采用创新的方法实时检测体内氧化还原参数。结果:利用高敏感的基因编码生物传感器HyPer7和光纤神经接口技术,我们发现在大脑中动脉闭塞后的40小时内,大鼠大脑缺血区神经元和星形胶质细胞中的过氧化氢(H2O2)水平缓慢升高;值得注意的是,在星形胶质细胞中,这一水平略高。清醒小鼠的拉曼显微光谱也揭示了光血栓形成引起的急性缺血时神经元和星形胶质细胞线粒体氧化还原的差异。星形胶质细胞在缺血1小时后表现出电子传递链(ETC)的过载,而神经元则没有表现出减少电子携带量的变化。创新和结论:结合新的体内方法可以详细描述氧化还原事件的时空分辨率。我们在体内证明了神经元和星形胶质细胞在受损脑区的氧化还原差异。急性缺血期星形细胞ETC的电子负荷升高,为超氧阴离子自由基(O2•-)的生成增加及其随后转化为其他活性物质提供了基础。然而,我们观察到在发病后期星形胶质细胞和神经元中H2O2浓度升高。在此期间,ETC没有表现出电子负载的增加,因此,H2O2生成的增加可能是二次氧化还原事件的现象。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Redox Differences Between Neurons and Astrocytes <i>In Vivo</i> in Ischemic Brain Tissues of Rodents.","authors":"Daria A Kotova, Aleksandra D Ivanova, Ilya V Kelmanson, Kseniia I Morozova, Yulia V Khramova, Maxim A Solotenkov, Evgeny A Stepanov, Aleksandr A Moshchenko, Alisa B Tiaglik, Anna A Fedotova, Anton V Zalygin, Vladimir A Oleinikov, Alexey G Katrukha, Alexey Semyanov, Vsevolod V Belousov, Andrei B Fedotov, Ilya V Fedotov, Nadezda A Brazhe, Dmitry S Bilan","doi":"10.1089/ars.2024.0876","DOIUrl":"10.1089/ars.2024.0876","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Reactive oxygen species (ROS) are considered to play a key damaging role in brain during the development of ischemic stroke. To clarify how different ROS contribute to ischemic pathogenesis, innovative approaches for real-time <i>in vivo</i> detection of redox parameters are necessary. <b><i>Results:</i></b> Using highly sensitive genetically encoded biosensor HyPer7 and a fiber-optic neurointerface technology, we demonstrated that the level of hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) slowly increases in neurons and astrocytes of the ischemic area of the rat brain after middle cerebral artery occlusion during next 40 h; notably, in astrocytes the level is somewhat higher. Raman microspectroscopy in awake mice also revealed redox differences between mitochondria of neurons and astrocytes during acute ischemia caused by photothrombosis. Astrocytes demonstrated the overloading of the electron transport chain (ETC) with electrons after 1 h of ischemia, whereas neurons do not demonstrate changes in the amount of reduced electron carries. <b><i>Innovation and Conclusion:</i></b> The combination of novel <i>in vivo</i> approaches allows to detail redox events with spatiotemporal resolution. We demonstrated redox difference between neurons and astrocytes in damaged brain areas <i>in vivo</i>. An elevated loading of astrocytic ETC with electrons during the acute ischemia phase provides basis for the increased generation of superoxide anion radical (O<sub>2</sub><sup>•-</sup>) with its following conversion to other reactive species. However, we observed increased H<sub>2</sub>O<sub>2</sub> concentrations in astrocytes and neurons at later pathogenesis stages. During this period, ETC did not demonstrate an elevated loading with electrons, and therefore, increased H<sub>2</sub>O<sub>2</sub> generation may be a phenomenon of secondary redox events. <i>Antioxid. Redox Signal.</i> 43, 272-287.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"272-287"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144092718","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
Small Molecules Targeting Non-Coding RNAs Regulating Ferroptosis: New Opportunities in Precision Cancer Therapy. 靶向非编码rna调控铁下垂的小分子:精准癌症治疗的新机遇。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-08-01 Epub Date: 2025-05-23 DOI: 10.1089/ars.2024.0807
Junjing Zhang, Yan Wang, Joshua S Fleishman, Weihua Zheng, Hongquan Wang, Fanyu Meng, Yumin Wang

Ferroptosis, a distinct form of regulated cell death (RCD), has emerged as a promising approach for cancer treatment owing to its potential to inhibit tumor malignancy. Research indicates that non-coding RNAs (ncRNAs) regulate ferroptosis susceptibility in cancer cells through epigenetic modifications. ncRNAs play essential roles in cancer initiation, metastasis, and drug resistance. Findings indicate that small-molecule compounds (SMCs) target ncRNAs to regulate ferroptosis, providing new opportunities for precision cancer therapy. Therefore, this review aims to elucidate current molecular mechanisms underlying ncRNA-mediated ferroptosis regulation in cancer and investigate the potential of SMCs as therapeutic agents to modulate this process, offering a new strategy for precision in cancer treatment. This review also summarizes the innovative strategy of targeting ncRNAs with SMCs, a therapeutic approach for regulating ferroptosis and transforming the landscape of cancer treatment. Overall, it highlights a novel strategy for cancer therapy by pharmacologically targeting the ncRNA-ferroptosis axis with SMCs. Antioxid. Redox Signal. 43, 345-362.

铁下垂是一种不同形式的调节细胞死亡(RCD),由于其抑制肿瘤恶性肿瘤的潜力,已成为一种有希望的癌症治疗方法。研究表明,非编码rna (ncRNAs)通过表观遗传修饰调节癌细胞铁下垂的易感性。ncRNAs在癌症的发生、转移和耐药过程中发挥重要作用。研究结果表明,小分子化合物(SMCs)靶向ncrna调控铁凋亡,为癌症的精准治疗提供了新的机会。因此,本综述旨在阐明ncrna介导的铁凋亡在癌症中的调控机制,并研究SMCs作为治疗剂调节这一过程的潜力,为癌症的精确治疗提供新的策略。本文还总结了SMCs靶向ncrna的创新策略,这是一种调节铁凋亡和改变癌症治疗格局的治疗方法。总的来说,它强调了一种新的癌症治疗策略,即通过SMCs从药理学上靶向ncRNA-ferroptosis轴。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Small Molecules Targeting Non-Coding RNAs Regulating Ferroptosis: New Opportunities in Precision Cancer Therapy.","authors":"Junjing Zhang, Yan Wang, Joshua S Fleishman, Weihua Zheng, Hongquan Wang, Fanyu Meng, Yumin Wang","doi":"10.1089/ars.2024.0807","DOIUrl":"10.1089/ars.2024.0807","url":null,"abstract":"<p><p>Ferroptosis, a distinct form of regulated cell death (RCD), has emerged as a promising approach for cancer treatment owing to its potential to inhibit tumor malignancy. Research indicates that non-coding RNAs (ncRNAs) regulate ferroptosis susceptibility in cancer cells through epigenetic modifications. ncRNAs play essential roles in cancer initiation, metastasis, and drug resistance. Findings indicate that small-molecule compounds (SMCs) target ncRNAs to regulate ferroptosis, providing new opportunities for precision cancer therapy. Therefore, this review aims to elucidate current molecular mechanisms underlying ncRNA-mediated ferroptosis regulation in cancer and investigate the potential of SMCs as therapeutic agents to modulate this process, offering a new strategy for precision in cancer treatment. This review also summarizes the innovative strategy of targeting ncRNAs with SMCs, a therapeutic approach for regulating ferroptosis and transforming the landscape of cancer treatment. Overall, it highlights a novel strategy for cancer therapy by pharmacologically targeting the ncRNA-ferroptosis axis with SMCs. <i>Antioxid. Redox Signal.</i> 43, 345-362.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"345-362"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144126181","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
Targeting NADPH Oxidase with APX-115: Suppression of Platelet Activation and Thrombotic Response. 用 APX-115 靶向 NADPH 氧化酶:抑制血小板活化和血栓反应
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-08-01 Epub Date: 2025-04-04 DOI: 10.1089/ars.2024.0695
Joara Jang, Hyunseong Yu, Eun Bee Oh, Ji Won Park, Solee Kim, Taeryeong Kim, Jisue Sohn, Bo-Ram Jin, Tong-Shin Chang

Aims: NADPH oxidase (NOX)-derived reactive oxygen species (ROS) are critical for platelet activation and thrombus formation. We hypothesized that inhibiting NOX-mediated ROS production with a pan-NOX inhibitor, APX-115, could effectively suppress platelet activation and thrombus formation, potentially serving as a novel antiplatelet therapeutic. This study aimed to explore the effects of APX-115 on human platelet functional responses and ROS-mediated signaling pathways. Results: APX-115 inhibited intracellular and extracellular ROS production in collagen-stimulated platelets, suppressing aggregation, P-selectin exposure, and ATP release. By preserving protein tyrosine phosphatase activity, APX-115 reduced tyrosine phosphorylation-dependent pathways inhibition, including spleen tyrosine kinase, LAT, Vav1, Bruton's tyrosine kinase, and phospholipase Cγ2, leading to decreased PKC activation and calcium mobilization. APX-115 also suppressed collagen-induced integrin αIIbβ3 activation, accompanied by elevated cGMP and vasodilator-stimulated phosphoprotein phosphorylation levels. In addition, APX-115 reduced p38 MAPK and ERK5 activation, leading to diminished phospholipase A2 phosphorylation, thromboxane production, and the exposure of procoagulant phosphatidylserine. These inhibitory effects extended to thrombus development caused by platelet adherence under shear and arterial thrombosis without prolonging bleeding time in murine models. Innovation: This study is the first to demonstrate that APX-115 inhibits NOX-mediated ROS production, platelet activation, and thrombus formation. By uncovering its effects on collagen receptor glycoprotein VI-mediated pathways, the work highlights the promise of APX-115 as an antiplatelet and antithrombotic agent. Conclusion: Our findings highlight the therapeutic potential of APX-115 in treating thrombotic and cardiovascular disorders by targeting NOX-mediated ROS production to mitigate platelet hyperreactivity and thrombus formation. Antioxid. Redox Signal. 43, 288-307.

目的:NADPH氧化酶(NOX)衍生的活性氧(ROS)对血小板活化和血栓形成至关重要。我们假设用泛氮氧化物抑制剂APX-115抑制氮氧化物介导的ROS产生,可以有效抑制血小板活化和血栓形成,可能作为一种新的抗血小板治疗药物。本研究旨在探讨APX-115对人血小板功能反应和ros介导的信号通路的影响。结果:APX-115抑制胶原刺激血小板细胞内和细胞外ROS的产生,抑制聚集、p -选择素暴露和ATP释放。通过保持蛋白酪氨酸磷酸酶活性,APX-115减少酪氨酸磷酸化依赖途径的抑制,包括脾酪氨酸激酶、LAT、Vav1、布鲁顿酪氨酸激酶和磷脂酶Cγ2,导致PKC活化和钙动员降低。APX-115还抑制胶原诱导的整合素α ib β3活化,并伴有cGMP和血管扩张剂刺激的磷酸化水平升高。此外,APX-115降低了p38 MAPK和ERK5的激活,导致磷脂酶A2磷酸化、血栓素产生和促凝磷脂酰丝氨酸暴露减少。在小鼠模型中,这些抑制作用扩展到剪切和动脉血栓形成下血小板粘附引起的血栓形成,而不延长出血时间。创新:本研究首次证明APX-115抑制nox介导的ROS生成、血小板活化和血栓形成。通过揭示其对胶原受体糖蛋白vi介导途径的影响,这项工作强调了APX-115作为抗血小板和抗血栓药物的前景。结论:我们的研究结果强调了APX-115通过靶向nox介导的ROS产生来减轻血小板高反应性和血栓形成,在治疗血栓和心血管疾病方面的治疗潜力。Antioxid。氧化还原信号:00000 - 00000。[图:见正文]。
{"title":"Targeting NADPH Oxidase with APX-115: Suppression of Platelet Activation and Thrombotic Response.","authors":"Joara Jang, Hyunseong Yu, Eun Bee Oh, Ji Won Park, Solee Kim, Taeryeong Kim, Jisue Sohn, Bo-Ram Jin, Tong-Shin Chang","doi":"10.1089/ars.2024.0695","DOIUrl":"10.1089/ars.2024.0695","url":null,"abstract":"<p><p><b><i>Aims:</i></b> NADPH oxidase (NOX)-derived reactive oxygen species (ROS) are critical for platelet activation and thrombus formation. We hypothesized that inhibiting NOX-mediated ROS production with a pan-NOX inhibitor, APX-115, could effectively suppress platelet activation and thrombus formation, potentially serving as a novel antiplatelet therapeutic. This study aimed to explore the effects of APX-115 on human platelet functional responses and ROS-mediated signaling pathways. <b><i>Results:</i></b> APX-115 inhibited intracellular and extracellular ROS production in collagen-stimulated platelets, suppressing aggregation, P-selectin exposure, and ATP release. By preserving protein tyrosine phosphatase activity, APX-115 reduced tyrosine phosphorylation-dependent pathways inhibition, including spleen tyrosine kinase, LAT, Vav1, Bruton's tyrosine kinase, and phospholipase Cγ2, leading to decreased PKC activation and calcium mobilization. APX-115 also suppressed collagen-induced integrin αIIbβ3 activation, accompanied by elevated cGMP and vasodilator-stimulated phosphoprotein phosphorylation levels. In addition, APX-115 reduced p38 MAPK and ERK5 activation, leading to diminished phospholipase A2 phosphorylation, thromboxane production, and the exposure of procoagulant phosphatidylserine. These inhibitory effects extended to thrombus development caused by platelet adherence under shear and arterial thrombosis without prolonging bleeding time in murine models. <b><i>Innovation:</i></b> This study is the first to demonstrate that APX-115 inhibits NOX-mediated ROS production, platelet activation, and thrombus formation. By uncovering its effects on collagen receptor glycoprotein VI-mediated pathways, the work highlights the promise of APX-115 as an antiplatelet and antithrombotic agent. <b><i>Conclusion:</i></b> Our findings highlight the therapeutic potential of APX-115 in treating thrombotic and cardiovascular disorders by targeting NOX-mediated ROS production to mitigate platelet hyperreactivity and thrombus formation. <i>Antioxid. Redox Signal.</i> 43, 288-307.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"288-307"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143778872","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
Unraveling Ammonia-Induced Brain Endothelial Senescence: Role of miRNA-183-5p. 揭示氨诱导的脑内皮衰老:miRNA-183-5p的作用。
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-08-01 Epub Date: 2025-05-23 DOI: 10.1089/ars.2024.0784
Karolina Orzeł-Gajowik, Krzysztof Milewski, Marta Obara-Michlewska, Aleksandra Ellert-Miklaszewska, Aneta Magiera, Karina Kwapiszewska, Magdalena Zielińska

Aims: Hyperammonemia, defined by elevated ammonia levels, may co-occur in various neurological disorders, but its effects on cerebrovascularity are not fully understood. This study aimed to investigate how hyperammonemia affects brain endothelial cells senescence and selected within in silico analysis micro RNA-183-5p in this process. Results: Reduction in cerebrovascular density in hyperammonemia-induced rats, similar to that seen in 12-month-old rats, using von Willebrand factor staining, was observed. MicroRNA (miRNA) profile analysis of the brain cortex and plasma identified miRNA-183-5p contributing to endothelial senescence. In vitro studies of ammonia-treated rat brain endothelial cell line 4 showed senescent features, including increased β-galactosidase activity, higher mRNA levels and fluorescence intensity of p16 and p21, and altered senescence-associated secretory phenotype. Additionally, the transfection of miRNA-183-5p mimic induced similar senescent characteristics in endothelial cells, whereas miRNA-183-5p mimic inhibition reversed some effects. Innovation: This study is the first to link hyperammonemia-induced cerebrovascular dysfunction with miRNA-183-5p, highlighting its role in promoting endothelial senescence. The findings suggest that miRNA-183-5p could be a target for therapeutic interventions, preventing ammonia-induced brain endothelial dysfunction. Conclusion: Hyperammonemia promotes brain endothelial cells senescence through miRNA-183-5p, reducing cerebrovascular density. This may contribute to cerebral dysfunction seen in hyperammonemia-associated neurological disorders. Targeting miRNA-183-5p could offer a novel therapeutic strategy to mitigate endothelial dysfunction and preserve brain health in hyperammonemia. Antioxid. Redox Signal. 43, 254-271.

目的:高氨血症,由氨水平升高定义,可能同时发生在各种神经系统疾病中,但其对脑血管的影响尚不完全清楚。本研究旨在探讨高氨血症对脑内皮细胞衰老的影响,并在此过程中筛选微RNA-183-5p。结果:采用血管性血友病因子染色,观察到高氨诱导大鼠脑血管密度降低,与12月龄大鼠相似。大脑皮层和血浆的MicroRNA (miRNA)谱分析发现,miRNA-183-5p与内皮细胞衰老有关。氨处理大鼠脑内皮细胞系4的体外研究显示出衰老特征,包括β-半乳糖苷酶活性升高,p16和p21 mRNA水平和荧光强度升高,衰老相关分泌表型改变。此外,转染miRNA-183-5p模拟物诱导内皮细胞类似的衰老特征,而miRNA-183-5p模拟物抑制逆转了一些作用。创新点:本研究首次将高氨血症引起的脑血管功能障碍与miRNA-183-5p联系起来,突出了其促进内皮细胞衰老的作用。研究结果表明,miRNA-183-5p可能是治疗干预的靶点,可以预防氨诱导的脑内皮功能障碍。结论:高氨血症通过miRNA-183-5p促进脑内皮细胞衰老,降低脑血管密度。这可能导致高氨相关神经系统疾病的脑功能障碍。靶向miRNA-183-5p可能提供一种新的治疗策略,以减轻高氨血症患者的内皮功能障碍和保持大脑健康。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Unraveling Ammonia-Induced Brain Endothelial Senescence: Role of miRNA-183-5p.","authors":"Karolina Orzeł-Gajowik, Krzysztof Milewski, Marta Obara-Michlewska, Aleksandra Ellert-Miklaszewska, Aneta Magiera, Karina Kwapiszewska, Magdalena Zielińska","doi":"10.1089/ars.2024.0784","DOIUrl":"10.1089/ars.2024.0784","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Hyperammonemia, defined by elevated ammonia levels, may co-occur in various neurological disorders, but its effects on cerebrovascularity are not fully understood. This study aimed to investigate how hyperammonemia affects brain endothelial cells senescence and selected within <i>in silico</i> analysis micro RNA-183-5p in this process. <b><i>Results:</i></b> Reduction in cerebrovascular density in hyperammonemia-induced rats, similar to that seen in 12-month-old rats, using von Willebrand factor staining, was observed. MicroRNA (miRNA) profile analysis of the brain cortex and plasma identified miRNA-183-5p contributing to endothelial senescence. <i>In vitro</i> studies of ammonia-treated rat brain endothelial cell line 4 showed senescent features, including increased β-galactosidase activity, higher mRNA levels and fluorescence intensity of p16 and p21, and altered senescence-associated secretory phenotype. Additionally, the transfection of miRNA-183-5p mimic induced similar senescent characteristics in endothelial cells, whereas miRNA-183-5p mimic inhibition reversed some effects. <b><i>Innovation:</i></b> This study is the first to link hyperammonemia-induced cerebrovascular dysfunction with miRNA-183-5p, highlighting its role in promoting endothelial senescence. The findings suggest that miRNA-183-5p could be a target for therapeutic interventions, preventing ammonia-induced brain endothelial dysfunction. <b><i>Conclusion:</i></b> Hyperammonemia promotes brain endothelial cells senescence through miRNA-183-5p, reducing cerebrovascular density. This may contribute to cerebral dysfunction seen in hyperammonemia-associated neurological disorders. Targeting miRNA-183-5p could offer a novel therapeutic strategy to mitigate endothelial dysfunction and preserve brain health in hyperammonemia. <i>Antioxid. Redox Signal.</i> 43, 254-271.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"254-271"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144126094","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
Downregulation of the Zinc Transporter ZIP13 (Slc39a13) Leads to Ferroptosis by Inhibiting Mitochondrial Iron-Sulfur Cluster Biosynthesis and Induces Ischemia/Reperfusion Injury in Mouse Hearts. 下调锌转运蛋白ZIP13 (Slc39a13)通过抑制线粒体铁硫团团生物合成导致铁凋亡并诱导小鼠心脏缺血/再灌注损伤
IF 6.1 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-08-01 Epub Date: 2025-05-19 DOI: 10.1089/ars.2024.0815
Rui Zhang, Jiannan Wang, Qing Yang, Yonghao Yu, Xinxin Cheng, Zhelong Xu

Aims: While ferroptosis is involved in the pathogenesis of myocardial ischemia/reperfusion (I/R) injury, the exact mechanism underlying the induction of ferroptosis by I/R remains elusive. Since downregulation of Zrt, Irt-like protein 13 (ZIP13) plays a role in I/R injury by targeting mitochondria, we hypothesized that ZIP13 downregulation during I/R leads to ferroptosis through a mitochondria-dependent mechanism. Results: ZIP13 cKO (cardiac-specific conditional knockout) induced ferroptosis and suppressed mitochondrial iron-sulfur cluster (ISC) biosynthesis. ZIP13 cKO also reduced glutathione levels as well as solute carrier family 7 member 11 (SLC7A11) expression. Moreover, cKO increased mitochondrial Fe2+ levels. Similar to the action of cKO, I/R led to ZIP13 downregulation, ferroptosis, mitochondrial Fe2+ accumulation, and suppression of ISC biosynthesis. In support, cKO of ZIP13 aggravated I/R-induced ferroptosis and mitochondrial Fe2+ accumulation. In contrast, ZIP13 overexpression prevented I/R-induced ferroptosis, mitochondrial Fe2+ accumulation, and suppression of ISC biosynthesis. Finally, ferrostatin-1, a ferroptosis inhibitor, alleviated I/R-induced ferroptosis as well as cardiac injury in cKO mice. Innovation: This study proposes a previously unknown mechanism by which ZIP13 downregulation contributes to ferroptosis in the setting of myocardial I/R. Conclusions: These findings highlight that ZIP13 downregulation at reperfusion triggers ferroptosis by suppressing the mitochondrial ISC biosynthesis followed by mitochondrial Fe2+ accumulation. Downregulation of SLC7A11 may also contribute to the action of ZIP13 downregulation. Antioxid. Redox Signal. 43, 328-344.

目的:虽然铁下垂参与心肌缺血/再灌注(I/R)损伤的发病机制,但I/R诱导铁下垂的确切机制尚不清楚。由于Zrt下调,irt样蛋白13 (ZIP13)通过靶向线粒体在I/R损伤中发挥作用,我们假设ZIP13在I/R过程中下调通过线粒体依赖机制导致铁凋亡。结果:ZIP13 cKO(心脏特异性条件敲除)诱导铁下垂并抑制线粒体铁硫团块(ISC)的生物合成。ZIP13 cKO还降低了谷胱甘肽水平和溶质载体家族7成员11 (SLC7A11)的表达。此外,cKO增加了线粒体Fe2+水平。与cKO的作用类似,I/R导致ZIP13下调、铁下垂、线粒体Fe2+积累和ISC生物合成抑制。ZIP13的cKO加重了I/ r诱导的铁下垂和线粒体Fe2+积累。相比之下,ZIP13过表达可阻止I/ r诱导的铁下垂、线粒体Fe2+积累和ISC生物合成的抑制。最后,铁抑制素-1 (ferrostatin-1)减轻了I/ r诱导的cKO小鼠铁下垂和心脏损伤。创新:本研究提出了一种以前未知的机制,即ZIP13下调有助于心肌I/R环境下的铁下垂。结论:这些研究结果表明,再灌注时ZIP13的下调通过抑制线粒体ISC的生物合成以及线粒体Fe2+的积累来触发铁下垂。SLC7A11的下调也可能导致ZIP13的下调。Antioxid。氧化还原信号:00000 - 00000。
{"title":"Downregulation of the Zinc Transporter ZIP13 (Slc39a13) Leads to Ferroptosis by Inhibiting Mitochondrial Iron-Sulfur Cluster Biosynthesis and Induces Ischemia/Reperfusion Injury in Mouse Hearts.","authors":"Rui Zhang, Jiannan Wang, Qing Yang, Yonghao Yu, Xinxin Cheng, Zhelong Xu","doi":"10.1089/ars.2024.0815","DOIUrl":"10.1089/ars.2024.0815","url":null,"abstract":"<p><p><b><i>Aims:</i></b> While ferroptosis is involved in the pathogenesis of myocardial ischemia/reperfusion (I/R) injury, the exact mechanism underlying the induction of ferroptosis by I/R remains elusive. Since downregulation of Zrt, Irt-like protein 13 (ZIP13) plays a role in I/R injury by targeting mitochondria, we hypothesized that ZIP13 downregulation during I/R leads to ferroptosis through a mitochondria-dependent mechanism. <b><i>Results:</i></b> ZIP13 cKO (cardiac-specific conditional knockout) induced ferroptosis and suppressed mitochondrial iron-sulfur cluster (ISC) biosynthesis. ZIP13 cKO also reduced glutathione levels as well as solute carrier family 7 member 11 (SLC7A11) expression. Moreover, cKO increased mitochondrial Fe<sup>2+</sup> levels. Similar to the action of cKO, I/R led to ZIP13 downregulation, ferroptosis, mitochondrial Fe<sup>2+</sup> accumulation, and suppression of ISC biosynthesis. In support, cKO of ZIP13 aggravated I/R-induced ferroptosis and mitochondrial Fe<sup>2+</sup> accumulation. In contrast, ZIP13 overexpression prevented I/R-induced ferroptosis, mitochondrial Fe<sup>2+</sup> accumulation, and suppression of ISC biosynthesis. Finally, ferrostatin-1, a ferroptosis inhibitor, alleviated I/R-induced ferroptosis as well as cardiac injury in cKO mice. <b><i>Innovation:</i></b> This study proposes a previously unknown mechanism by which ZIP13 downregulation contributes to ferroptosis in the setting of myocardial I/R. <b><i>Conclusions:</i></b> These findings highlight that ZIP13 downregulation at reperfusion triggers ferroptosis by suppressing the mitochondrial ISC biosynthesis followed by mitochondrial Fe<sup>2+</sup> accumulation. Downregulation of SLC7A11 may also contribute to the action of ZIP13 downregulation. <i>Antioxid. Redox Signal.</i> 43, 328-344.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"328-344"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144101025","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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

Aims: 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. Results: 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. Innovation: 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. Conclusions: 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. Antioxid. Redox Signal. 43, 138-150.

目的:帕金森病(PD)以黑质致密部(SNpc)多巴胺能(DAergic)神经元变性为特征。硫氧还蛋白-1 (Trx-1)是一种氧化还原蛋白,可保护神经元免受各种损伤。我们的研究表明,Trx-1过表达可改善1-甲基-4-苯基-1,2,3,6-四氢吡啶(MPTP)诱导的学习和记忆障碍。然而,Trx-1调控的SNpc向海马的特异性信号传递在PD相关认知缺陷中的作用尚不清楚。结果:我们发现Trx-1在SNpc中的下调加重了MPTP诱导的认知功能障碍。重要的是,我们观察到SNpc直接投射到海马体。我们发现MPTP诱导SNpc中DAergic神经元的缺失导致海马多巴胺D1受体(D1R)表达减少,这是由SNpc中Trx-1下调促进的。MPTP降低海马中磷酸化细胞外信号调节激酶(p-ERK1/2)、磷酸化camp反应元件结合蛋白(p-CREB)、脑源性神经营养因子(BDNF)和突触后密度蛋白95 (PSD95)的水平,并通过SNpc中Trx-1的下调进一步降低。最后,海马MPTP减少海马突触数量,SNpc下调Trx-1进一步减少突触数量。创新点:Trx-1下调加速SNpc中DAergic神经元的丢失,导致多巴胺能向海马的预测数量减少,从而抑制海马中的D1R-ERK1/2-CREB-BDNF通路,最终损害海马依赖性认知。结论:这些结果表明SNpc中Trx-1水平的降低通过降低海马D1R信号通路在PD个体认知功能障碍中起重要的调节作用。Antioxid。氧化还原信号:00000 - 00000。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

Significance: 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. Recent Advances: 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. Critical Issues: 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+)/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. Future Directions: 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. Antioxid. Redox Signal. 43, 92-114.

意义:本综述探讨放射治疗(RT)如何增加癌症幸存者迟发性心血管疾病(CVD)的风险。了解辐射诱发CVD的机制对于开发靶向治疗以减轻这些影响和改善癌症患者的长期预后至关重要。最新进展:最近的研究主要集中在辐射引起的各种癌症细胞类型的代谢改变。然而,关于正常细胞,特别是血管细胞的慢性代谢改变在rt后CVD进展中的作用,仍然存在显著的知识差距。关键问题:本综述集中于rt诱导的血管细胞代谢改变及其对rt后血管系统衰老积累和慢性炎症的贡献。我们讨论了关键的代谢途径,包括糖酵解、三羧酸循环、脂质代谢、谷氨酰胺代谢和氧化还原代谢(烟酰胺腺嘌呤二核苷酸/烟酰胺腺嘌呤二核苷酸(NADH)和烟酰胺腺嘌呤二核苷酸磷酸(NADP+)/NADPH)。我们进一步探讨了p53、单磷酸腺苷活化蛋白激酶和哺乳动物雷帕霉素靶蛋白等调节蛋白在驱动这些代谢失调中的作用。这篇综述强调了由衰老相关分泌表型介导的免疫-血管串扰的影响,它使代谢功能障碍永久化,增强慢性炎症,促进衰老积累,并导致血管损伤,最终导致心血管发病。未来方向:未来的研究应优先确定这些代谢途径中的治疗靶点或受rt影响的免疫血管相互作用。通过靶向治疗纠正代谢功能障碍和减少慢性炎症可以显著改善癌症幸存者的心血管预后。Antioxid。氧化还原信号:00000 - 00000。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

Aims: 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. Results: 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. In vitro, 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. Innovation and Conclusion: 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. Antioxid. Redox Signal. 43, 56-74.

目的:高尿酸血症肾病(HN)是高尿酸血症的一种常见并发症,主要表现为肾小管功能障碍、炎症和进行性肾纤维化,其发病机制尚不清楚。铁蛋白沉积是一种铁依赖性调控细胞死亡,与多种疾病有关,但很少与 HN 联系在一起。在本研究中,我们旨在探讨铁蛋白沉积在 HN 中的可能作用及其潜在机制。结果显示我们发现尿酸氧化酶基因敲除小鼠(一种高尿酸血症模型)表现出肾功能损害,尿酸、肌酐和血尿素氮水平升高,同时伴有铁沉积增加、谷胱甘肽过氧化物酶 4 (GPX4) 和 xCT 表达减少,这表明铁氧化参与其中。铁变态反应抑制剂铁前列素-1(Fer-1)可改善这些小鼠的肾损伤、炎症细胞浸润和纤维化。从机理上讲,Fer-1 恢复了抗氧化蛋白水平,使铁蛋白沉积相关蛋白表达正常化,减轻了铁过载和脂质过氧化,抑制了炎症标志物和丝裂原活化蛋白激酶信号传导。在体外,单钠尿酸盐结晶诱导人肾 2 细胞发生铁变态反应,其特征是脂质过氧化和铁积累增加。值得注意的是,抑制高级糖化终产物受体(RAGE)可减轻肾损伤、炎症和纤维化,但不会直接减轻铁沉积。这些发现在人类高尿酸血症相关肾病样本中得到了验证,这些样本显示铁沉积增加、GPX4 减少和 RAGE 表达升高。创新与结论:本研究表明,铁蛋白沉积可能在高尿酸血症肾损伤、炎症和纤维化的发展过程中发挥作用,并可能通过 RAGE 信号传导。虽然抑制 RAGE 可改善肾损伤,但它并不直接影响铁蛋白沉积,这表明 RAGE 在肾损伤中的作用是复杂的,且取决于具体情况。这些发现凸显了铁蛋白沉积及其相关途径(包括 RAGE 信号转导)是 HN 的潜在治疗靶点。抗氧化。氧化还原信号。00, 000-000.
{"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
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