{"title":"Peroxiredoxin 6: A Regulatory Target in Cellular Senescence and Age-Related Diseases.","authors":"Hao Wang, Yingjie Zhao, Fuli Zhou, Fan Chen, Tianyu Chen, Jinhong Wang, Hao Liu, Cheng Sun, Renpeng Zhou, Wei Hu, Chao Lu","doi":"10.1089/ars.2024.0793","DOIUrl":"10.1089/ars.2024.0793","url":null,"abstract":"<p><p><b><i>Significance:</i></b> Oxidative stress is a key factor in inducing cellular senescence and age-related diseases. When the redox balance within the body is disrupted, sustained oxidative stress can lead to cellular senescence. The accumulation of senescent cells, in turn, triggers a variety of age-related diseases. Peroxiredoxin 6 (Prdx6) is a critical target in the intracellular regulation of redox homeostasis. <b><i>Recent Advances:</i></b> Prdx6 is a widely expressed antioxidant enzyme and the sole member of the peroxiredoxin family endowed with multiple enzymatic functions, including peroxidase activity along with acidic calcium-independent phospholipase A2 (aiPLA2) and lysophosphatidylcholine acyltransferase (LPCAT) activities. Its fundamental physiological functions involve protecting against oxidative stress and maintaining phospholipid homeostasis. Recent studies have shown that Prdx6 is widely involved in the regulation of cellular senescence and influences the development and progression of various age-related diseases. <b><i>Critical Issues:</i></b> Cellular senescence and age-related diseases, due to their complex mechanisms, lack effective treatments. Therefore, there is an urgent need to identify new therapeutic targets. This review discusses the biological characteristics of Prdx6 and its pathophysiological roles in cellular senescence and age-related diseases. Prdx6 may serve as a potential target for modulating cellular senescence and age-related diseases. <b><i>Future Directions:</i></b> The regulatory mechanisms of Prdx6 in age-related diseases warrant further investigation. Additionally, conducting drug screening to identify more molecules that can specifically target Prdx6 will provide new strategies for the treatment of age-related diseases. <i>Antioxid. Redox Signal.</i> 43, 400-426.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"400-426"},"PeriodicalIF":6.1,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144265070","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}
{"title":"Uric Acid Stimulates PINK1/Parkin-Mediated Mitophagy via Nrf2/HO-1 Pathway to Protect Against Neuronal Apoptosis in Alzheimer's Disease.","authors":"Qian Zhang, De Xie, Binyang Chen, Linqian Yu, Jiayu Chen, Yunbo Yan, Mingyan Zhang, Qiang Wang, Yuemei Xi, Tetsuya Yamamoto, Hidenori Koyama, Jidong Cheng","doi":"10.1089/ars.2024.0837","DOIUrl":"10.1089/ars.2024.0837","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder among the elderly. Uric acid (UA), the end product of purine metabolism, functions as a potent free radical scavenger and helps mitigate oxidative stress. Several epidemiological studies revealed that serum UA levels are negatively correlated with the risk of AD; however, the molecular mechanisms remain unclear. Notably, β-amyloid (Aβ) deposition is implicated in the disruption of mitophagy, leading to neuronal apoptosis. In this study, we aim to elucidate the link between UA and AD and explore the underlying mechanisms. <b><i>Results:</i></b> We demonstrated that UA improved cognitive impairment in 5×FAD mice and reduced neuronal apoptosis both <i>in vivo</i> and <i>in vitro</i>. UA reversed the expression of phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1), p-Parkin<sup>S65</sup>, parkin, microtubule-associated protein 1 light chain 3 II/I, and p62 proteins inhibited by Aβ treatment, alleviated Aβ induced mitochondrial dysfunction, and disturbed dynamics. We found that UA activated nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1(HO-1) signaling both <i>in vivo</i> and <i>in vitro</i>. Furthermore, ML385, a Nrf2-specific inhibitor, reversed the increase in mitochondrial membrane potential and mitophagy promoted by UA and increased neuronal apoptosis in HT22 cells. The antiapoptotic effects of UA in HT22 cells were prevented by treatment with small interfering RNAs targeting PINK1. <b><i>Conclusions and Innovation:</i></b> These data suggest that UA stimulates PINK1/parkin-mediated mitophagy reducing Aβ-induced neuronal apoptosis through the Nrf2/HO-1 pathway, which plays a neuroprotective role in AD. Our findings confirmed that UA effectively reduces neuronal damage and cognitive impairment, highlighting its potential clinical applications in the treatment of AD. <i>Antioxid. Redox Signal.</i> 43, 381-399. [Figure: see text].</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"381-399"},"PeriodicalIF":6.1,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144504732","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}
Chien-Lin Lu, Yi-Shiou Tseng, Wen-Bin Wu, Chun-Hou Liao, Ming-Chieh Ma
{"title":"Blockade of Aryl Hydrocarbon Receptor Ameliorates Functional Insufficiency in 5/6 Nephrectomized Rat Kidneys by Restoring Hydrogen Sulfide Formation.","authors":"Chien-Lin Lu, Yi-Shiou Tseng, Wen-Bin Wu, Chun-Hou Liao, Ming-Chieh Ma","doi":"10.1089/ars.2024.0833","DOIUrl":"10.1089/ars.2024.0833","url":null,"abstract":"<p><p><b><i>Aims:</i></b> We previously demonstrated that aryl hydrocarbon receptor (AhR) activation attenuates the cytoprotective effect of hydrogen sulfide (H<sub>2</sub>S), leading to indoxyl sulfate (IS)-mediated renal tubular damage. However, it is unclear whether this pathway would be present in an <i>in vivo</i> uremic model. <b><i>Results:</i></b> In a rat chronic kidney disease (CKD) model with 5/6 nephrectomized (Nx), we found that poor renal filtration is associated with accumulation of IS and homocysteine (Hcy), an H<sub>2</sub>S precursor. Compared with controls, the protein and mRNA levels of H<sub>2</sub>S-producing enzymes, including cystathionine β-synthase, cystathionine γ-lyase, and 3-mercaptopyruvate sulfurtransferase, were attenuated in Nx kidneys. Since the transcription factor, specificity protein 1 (Sp1), acts as an upstream regulator of these enzyme expressions, we found that the protein level and activity of Sp1 were significantly decreased in Nx kidneys. Interestingly, employing the blocker of the AhR CH-223191 not only reverses the decrease in H<sub>2</sub>S-producing enzymes and Sp1, but it also reverses H<sub>2</sub>S reduction in Nx rats. These are associated with the mitigation of plasma Hcy accumulation, renal excretion, perfusion insufficiency, and tubular damage. Moreover, the oxidative stress in Nx kidneys due to increased superoxide formation and decreased glutathione contents was also attenuated by AhR inhibition. <b><i>Innovation:</i></b> Our findings highlight the deleterious effect of AhR activation on renal H<sub>2</sub>S formation may be due to IS accumulation and underline AhR blockade as a novel therapy for CKD. <b><i>Conclusion:</i></b> AhR is detrimental to Sp1 function <i>in vivo</i>, leading to impeding renal H<sub>2</sub>S generation and exacerbating oxidative stress during CKD progression. <i>Antioxid. Redox Signal.</i> 43, 448-464.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"448-464"},"PeriodicalIF":6.1,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144574708","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}
{"title":"Combined Deletion of Mitofusin 2 in Adipose-Mesenchymal Derived Stem Cells and Melatonin Offers Additional Benefits on Protecting the Brain Against Acute Ischemic Stroke in Rat.","authors":"Chien-Hui Yang, Hung-Sheng Lin, Han-Tan Chai, Yi-Ling Chen, Hon-Kan Yip, Kuan-Hung Chen","doi":"10.1177/15230864251364881","DOIUrl":"10.1177/15230864251364881","url":null,"abstract":"<p><p><b><i>Background and Aims:</i></b> Ischemic stroke (IS) remains the third leading cause of death, and the treatment of acute ischemic stroke (AIS) is still a formidable challenge to clinicians. This study tested the hypothesis that combined silencing Mnf2 gene in adipose-derived mesenchymal stem cells (ADMSCs<sup>sil-Mnf2</sup>) and melatonin (Mel) therapy was superior to monotherapy on attenuating the brain infarct volume (BIV) and improving neurological function in AIS rats. <b><i>Results:</i></b> <i>In vitro</i> and <i>in vivo</i> studies were conducted. <i>In vitro</i> results showed that as compared with the controls (<i>i.e.,</i> ADMSCs/N2a cells), the cellular/protein levels of oxidative stress/reactive oxygen species (ROS)/mitochondrial and DNA damaged/apoptotic/cell stress signaling (tumor necrosis factor [TNF] receptor associated factor 6/ apoptosis signal regulating kinase/MKK<sup>4/7</sup>/JUN/ERK<sup>1/2</sup>/c-Jun) biomarkers were significantly increased in these cells treated by H<sub>2</sub>O<sub>2</sub> that were significantly reversed by ADMSCs<sup>sil-Mnf2</sup> or Mel and further significantly reversed by combined therapy (all <i>p</i> < 0.0001). Animals were categorized into groups 1 (sham-operated control)/2 (AIS)/3 (AIS + Mel)/4 (AIS + ADMSCs<sup>sil-Mnf2</sup>)/5 (AIS + Mel-ADMSCs<sup>sil-Mnf2</sup>) and euthanized by day 28 after AIS. By day 28, the BIV and the brain infarct area (BIA) were lowest in group 1/highest in group 2/significantly lower in group 5 than in groups 3 and 4/significantly increased in group 4 than in group 3, whereas the neurological function displayed an opposite manner of BIV (all <i>p</i> < 0.0001). The protein expressions of oxidative stress/mitochondrial damaged/apoptotic/inflammatory/cell stress signaling biomarkers displayed an identical pattern, whereas the protein expressions of mitochondrial biogenesis/antioxidants and cellular level of neuronal cells exhibited an opposite manner of BIV among the groups (all <i>p</i> < 0.0001). <b><i>Innovation and Conclusion:</i></b> ADMSCs<sup>sil-Mnf2</sup> and Mel combined therapy offered synergic effects on attenuating the BIV/BIA and preserving neurological function in rodents after AIS mainly through suppressing oxidative stress/ROS/inflammatory signalings and upregulating antioxidants. Combined ADMSCs<sup>sil-Mnf2</sup> and Mel therapy offered additional benefits on protecting the brain against AIS in rodents. <i>Antioxid. Redox Signal.</i> 43, 427-447.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"427-447"},"PeriodicalIF":6.1,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144854281","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}
{"title":"Total Flavones of <i>Rhododendron</i> Promotes Microglial Polarization to the M2 Subtype via Inhibiting the NOX2/ROS Pathway in Poststroke Mice with Depression-Like Behavior.","authors":"Guoyi Deng, Zhifeng Geng, Xin Xu, Ziyu Wang, Xiaojiao Yin, Chenjing Hu, Zhiwu Chen, Jiyue Wen","doi":"10.1089/ars.2025.0948","DOIUrl":"10.1089/ars.2025.0948","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Total flavones of <i>Rhododendron</i> (TFR) extracted from the flowers of <i>Rhododendron</i> contains bioactive components. We investigated the main components of TFR and explored the role of TFR in microglial polarization in poststroke mice with depression-like behavior. <b><i>Results:</i></b> Using ultraperformance liquid chromatography-tandem mass spectrometry, we identified the main and potential active compounds in TFR as kaempferol, astragalin, epicatechin, myricetin, rutin, isoquercitrin, quercetin, and quercitrin. In addition, we demonstrated that TFR (60 and 120 mg/kg) efficiently ameliorated depression-like behavior in mice and promoted microglial polarization to the M2 subtype in the mouse hippocampal tissues. We also revealed that TFR (160 mg/L) facilitated microglial polarization to the M2 subtype following oxygen-glucose deprivation/reoxygenation (OGD/R) <i>in vitro</i>. RNA sequencing revealed the upregulation of NADPH oxidase 2 (NOX2) in the mouse brain tissues after cerebral ischemia/reperfusion (I/R) injury. TFR (120 mg/kg) inhibited NOX2 expression in the hippocampal tissues of cerebral I/R mice. In addition, TFR (160 mg/L) downregulated NOX2 expression in OGD/R-treated microglial cells and decreased reactive oxygen species (ROS) production. Notably, NOX2 inhibition promoted microglial polarization to the M2 subtype. <b><i>Conclusion and Innovation:</i></b> TFR promotes microglial polarization to the M2 subtype by inhibiting the NOX2/ROS pathway. <i>Antioxid. Redox Signal.</i> 43, 465-481.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"465-481"},"PeriodicalIF":6.1,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144493726","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}
Xingang Sun, Lu Chen, Jie Han, Weixun Cai, Shan Li, Ting Chen, Miao Chen, Han Zhang, Yuxian He, Liangrong Zheng, Lihong Wang
{"title":"ALOX15 Modulates Ferroptosis via the Reactive Oxygen Species-Mediated MAPK Pathway in Doxorubicin-Induced Cardiotoxicity.","authors":"Xingang Sun, Lu Chen, Jie Han, Weixun Cai, Shan Li, Ting Chen, Miao Chen, Han Zhang, Yuxian He, Liangrong Zheng, Lihong Wang","doi":"10.1089/ars.2023.0468","DOIUrl":"10.1089/ars.2023.0468","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Doxorubicin (Dox) is a potent chemotherapy agent, yet its clinical use is hampered by cardiotoxicity. Although extensive research has focused on Dox-induced cardiotoxicity (DIC), its mechanism remains elusive. Recent evidence implicates ferroptosis as a key contributor to DIC. The 15-lipoxygenase-1 (ALOX15), involved in lipid peroxidation, is known to play an essential role in ischemia-induced myocardial damage and heart failure; however, its function in DIC is undefined. This study seeks to elucidate the role of ALOX15 in DIC and unravel its underlying mechanism. <b><i>Results:</i></b> Both ALOX15 mRNA and protein levels were elevated in DIC models <i>in vivo</i> and <i>in vitro</i>. Inhibition or silencing of ALOX15 ameliorated lipid peroxidation, ferroptosis, and cardiac dysfunction in Dox-treated mice. Consistently, ALOX15 loss of function protected H9C2 cells against Dox and RSL3-induced toxicity. In addition, we found that linoleic acid increased the susceptibility of H9C2 cells toward Dox-induced damage, which was abolished by ALOX15 inhibition. Furthermore, <i>Alox15</i> overexpression aggravated Dox-induced cell damage by aggravating reactive oxygen species (ROS)-mediated ferroptosis. Mechanistically, we discovered that the amelioration of Dox-induced ferroptosis by ALOX15 loss of function occurred through inhibiting the ROS-mediated mitogen-activated protein kinase (MAPK) signaling pathway activation. <b><i>Innovation and Conclusion:</i></b> These results reveal that ALOX15 regulates ferroptosis through ROS-mediated MAPK signaling pathway in DIC, suggesting a potential therapeutic target for DIC intervention. <i>Antioxid. Redox Signal.</i> 43, 363-380.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"363-380"},"PeriodicalIF":6.1,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144207422","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}
{"title":"Reprogramming Iron Metabolism via the RIG-I/c-Myc/FTH Axis Mitigates Renal Ischemia-Reperfusion Injury.","authors":"Yulu Zhang, Jia Xing, Li Yao, Yu Zou, Hui Peng, Xiling Yi, Lifang Bai, Yang Yu, Hanzhe Liu, Xue Li, Xiaoyue Zhai","doi":"10.1177/15230864251369883","DOIUrl":"https://doi.org/10.1177/15230864251369883","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Iron metabolism disorders are critical in the pathogenesis of acute kidney ischemia-reperfusion injury (IRI). However, the molecular mechanisms driving these disturbances remain poorly understood. <b><i>Results:</i></b> In IRI mouse kidneys, pathological alterations, iron metabolism disruptions, and functional impairments were observed. Retinoic acid-inducible gene-I (RIG-I), transcription factor c-Myc, and ferritin heavy chain (FTH) exhibited elevated expression and colocalization in tubular epithelial cells, accompanied by decreased glutathione peroxidase 4 (GPX4) level and evidence of ferroptosis. Further <i>in vitro</i> studies revealed that RIG-I promoted c-Myc activation. The latter demonstrated its positive regulation of FTH transcription by chromatin immunoprecipitation assays and c-Myc siRNA experiments. Interestingly, FTH overexpression resulted in elevated levels of RIG-I, transferrin receptor, ferroportin, and nuclear receptor coactivator 4. Ultimately, the c-Myc inhibitor 10058-F4 reversed all adverse alterations and demonstrated a protective role in IRI mouse kidneys and mouse kidney tubule cells subjected to the ferroptosis inducer erastin, RIG-I agonist, or hypoxia/reoxygenation. This reversal was reflected in improved renal morphology and function, balanced iron metabolism, increased GPX4 level, decreased 4-hydroxynonenal level, reduced inflammatory cell infiltration, interleukin-1 beta release, and kidney injury molecule 1 expression. <b><i>Innovation:</i></b> This study proposes a novel mechanism in which c-Myc is activated by elevated RIG-I in IRI kidneys and positively regulates FTH transcription, therefore involving iron metabolism disorders. <b><i>Conclusions:</i></b> The RIG-I, c-Myc, and FTH disrupt iron homeostasis, and the c-Myc inhibition stabilizes iron metabolism and mitigates oxidative stress, suggesting a potential therapeutic target in IRI. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144939846","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Melatonin Ameliorates Cognitive Impairment Following Exertional Heat Stroke by Inhibiting Ferroptosis and Neuroinflammation.","authors":"Xiaochen Wang, Ziwei Han, Chao Liu, Jiaona Liu, Zhi Dai, Jie Hu, Zhi Mao, Qinglin Li, Xin Hu, Feihu Zhou","doi":"10.1177/15230864251363577","DOIUrl":"https://doi.org/10.1177/15230864251363577","url":null,"abstract":"<p><p><b><i>Aims:</i></b> This study aims to investigate whether melatonin (MLT) exerts protective effects against cognitive impairment following exertional heat stroke (EHS) by modulating ferroportin (Fpn) to alleviate hippocampal ferroptosis and neuroinflammation. <b><i>Results:</i></b> Following EHS, genes such as <i>Mt1</i>, <i>Mt2</i>, and <i>Trf</i> were notably upregulated in the hippocampal tissue, whereas genes such as <i>Slc40a1</i> (encoding Fpn 1) and <i>Il33</i> were downregulated. Kyoto Encyclopedia of Genes and Genomes analysis implicated ferroptosis as a dominant. MLT significantly ameliorated learning and memory deficits observed in EHS mice. This treatment also modulated ferroptosis markers, such as Fpn, xCT, ferritin H, and glutathione peroxidase 4, reduced hippocampal iron overload, and decreased the secretion of proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor-α (TNF-α). Furthermore, MLT treatment reduced oxidative stress and lipid peroxidation and mitigated mitochondrial and neuronal damage in the hippocampal tissue. Strikingly, conditional Fpn knockout abolished MLT's benefits: Fpn-cKO + MLT mice showed persistent iron accumulation, elevated IL-6 and TNF-α, and failed cognitive recovery. <b><i>Innovation:</i></b> Our study reveals that MLT prevents EHS-induced neurodegeneration by enhancing Fpn-dependent iron efflux, a mechanism that concurrently resolves hippocampal iron overload, suppresses ferroptosis, and dampens neuroinflammation. <b><i>Conclusion:</i></b> Our findings indicate that MLT mitigates EHS-related cognitive impairment by restoring hippocampal iron homeostasis and suppressing neuroinflammation, primarily through Fpn-dependent mechanisms. <i>Antioxid. Redox Signal.</i> 00, 000-000.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144803274","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Titanium Particles Activate Osteocytic Connexin 43 to Induce Oxidative Stress and Osteoclastogenesis Through the JAK-STAT Pathway.","authors":"Jiawei Ouyang, Hao Chai, Chunguang Sun, Shendong Wang, Chang She, Dechun Geng, Wei Xu","doi":"10.1089/ars.2024.0894","DOIUrl":"10.1089/ars.2024.0894","url":null,"abstract":"<p><p><b><i>Aims:</i></b> Periprosthetic osteolysis (PPO), a leading cause of aseptic loosening in joint replacement, arose from complex interactions among osteoblasts, osteoclasts, and osteocytes. Given the pivotal role of connexin 43 (Cx43) in osteocyte communication and bone remodeling, investigating its function was essential for understanding the mechanisms of osteolysis. Our previous studies showed that titanium (Ti) particles increased Cx43 expression in osteocytes. However, the role of Cx43 in osteolysis remained unclear. This study investigated the role of Cx43-mediated regulation of osteocytes on osteoclastogenesis in wear debris-induced osteolysis. <b><i>Results:</i></b> Using <i>Dmp1</i>-cre conditional <i>Cx43</i> knockout mice and the MLO-Y4 osteocyte cell line, we demonstrated that <i>Cx43</i> deficiency reduced bone resorption and osteoclastogenesis, thereby improving bone remodeling in a Ti particle-induced osteolysis model. Sequencing analysis revealed that <i>Cx43</i> gene expression changes might be linked to oxidative stress and the Janus Kinase (JAK)-STAT pathway. Elevated Cx43 expression in osteocytes stimulated by Ti particles increased STAT1 protein phosphorylation, induced oxidative stress, elevated the Receptor Activator of Nuclear Factor Kappa-Β Ligand (RANKL)/Osteoprotegerin (OPG) ratio, and promoted osteoclast activation and bone resorption. Conversely, <i>Cx43 gene</i> knockout decreased STAT1 protein phosphorylation and enhanced Nuclear Factor Erythroid 2-Related Factor 2 (NrF2) protein expression. Blocking the JAK-STAT signaling pathway activated by Cx43 increased NrF2 expression, reduced reactive oxygen species levels, and subsequently decreased the RANKL/OPG ratio. <b><i>Innovation and Conclusions:</i></b> This study identified a novel mechanism where Cx43 in osteocytes promoted osteoclastogenesis through JAK-STAT pathway activation and oxidative stress in wear debris-induced osteolysis. These findings highlighted the critical role of Cx43 in bone resorption and suggested targeting Cx43 or the JAK-STAT pathway as potential therapeutic strategies to mitigate osteolysis and improve implant longevity. <i>Antioxid. Redox Signal.</i> 43, 215-238.</p>","PeriodicalId":8011,"journal":{"name":"Antioxidants & redox signaling","volume":" ","pages":"215-238"},"PeriodicalIF":6.1,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143963209","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}