Lanlan Wang, Xuefei Dong, Luyao Yu, Haipeng Jie, Boyang Wang, Lei Li, Jing Chen, Meiyan Liu, Bo Dong
Diabetic cardiomyopathy (DCM) is a major complication of diabetes and a leading contributor to heart failure, in which cardiomyocyte senescence plays an increasingly recognized role. However, the underlying mechanisms driving this process remain poorly defined. Here, we identify the (pro)renin receptor (PRR) as a critical mediator of cardiomyocyte senescence in DCM. Using a high-fat diet and streptozotocin (STZ)-induced DCM mouse model, as well as primary cardiomyocytes exposed to high glucose and palmitic acid, we demonstrate that PRR expression is significantly upregulated in diabetic hearts and closely associated with key senescence markers, including SA-β-gal, γ-H2AX, p16, and p21. PRR overexpression exacerbates these senescence phenotypes and promotes the secretion of profibrotic senescence-associated secretory phenotype factors, contributing to increased myocardial fibrosis and cardiac dysfunction. Mechanistically, PRR stabilizes the p53 protein by inhibiting tripartite motif-containing 24 (TRIM24)-mediated ubiquitination and proteasomal degradation, thereby activating the p53-p21 axis. These findings reveal a novel role of the PRR in diabetic myocardial senescence and provide potential therapeutic targets for attenuating DCM progression.
{"title":"(Pro)renin receptor promotes cardiomyocyte senescence via tripartite motif-containing 24-mediated stabilization of p53 in diabetic cardiomyopathy.","authors":"Lanlan Wang, Xuefei Dong, Luyao Yu, Haipeng Jie, Boyang Wang, Lei Li, Jing Chen, Meiyan Liu, Bo Dong","doi":"10.3724/abbs.2025185","DOIUrl":"https://doi.org/10.3724/abbs.2025185","url":null,"abstract":"<p><p>Diabetic cardiomyopathy (DCM) is a major complication of diabetes and a leading contributor to heart failure, in which cardiomyocyte senescence plays an increasingly recognized role. However, the underlying mechanisms driving this process remain poorly defined. Here, we identify the (pro)renin receptor (PRR) as a critical mediator of cardiomyocyte senescence in DCM. Using a high-fat diet and streptozotocin (STZ)-induced DCM mouse model, as well as primary cardiomyocytes exposed to high glucose and palmitic acid, we demonstrate that PRR expression is significantly upregulated in diabetic hearts and closely associated with key senescence markers, including SA-β-gal, γ-H2AX, p16, and p21. PRR overexpression exacerbates these senescence phenotypes and promotes the secretion of profibrotic senescence-associated secretory phenotype factors, contributing to increased myocardial fibrosis and cardiac dysfunction. Mechanistically, PRR stabilizes the p53 protein by inhibiting tripartite motif-containing 24 (TRIM24)-mediated ubiquitination and proteasomal degradation, thereby activating the p53-p21 axis. These findings reveal a novel role of the PRR in diabetic myocardial senescence and provide potential therapeutic targets for attenuating DCM progression.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145761902","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}
Mengke He, Ningzhe Li, Yiwen Cao, Jie Jiang, Fan Zhang, Zeyi Li, Jie Shen, Dehao Zhu, Xiaxin Liu, Qiang Wang, Chenjing Ye, Junmin Li, Zhen Jin, Rufang Xiang
CD19-directed chimeric antigen receptor T (CAR-T) cell therapy is promising for treating relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL), yet its long-term efficacy remains limited by CAR-T-cell exhaustion. Given the broad immunomodulatory activities of vitamin D, we investigate whether its active form, 1α,25(OH) 2D 3, enhances CAR-T-cell functionality and improves therapeutic outcomes. We demonstrate that 1α,25(OH) 2D 3 treatment significantly mitigates exhaustion and enhances the antitumor activity of CD19 CAR-T cells derived from both healthy donors and DLBCL patients, which is further validated in xenograft mouse models. Mechanistically, we show that 1α,25(OH) 2D 3 upregulates the expression of the vitamin D receptor (VDR), promoting transcriptional reprogramming associated with memory-like differentiation and downregulation of exhaustion-related genes, thereby reshaping the functional heterogeneity of CAR-T cells under tumor stimulation. Our study highlights 1α,25(OH) 2D 3 supplementation as a safe and accessible approach to mitigate terminal differentiation and exhaustion of CAR-T cells, offering a promising strategy to enhance the clinical efficacy of CAR-T therapy in patients with R/R DLBCL.
{"title":"1α,25(OH) <sub>2</sub>D <sub>3</sub> prevents CD19 CAR-T cell exhaustion and differentiation via VDR-dependent transcriptional reprogramming.","authors":"Mengke He, Ningzhe Li, Yiwen Cao, Jie Jiang, Fan Zhang, Zeyi Li, Jie Shen, Dehao Zhu, Xiaxin Liu, Qiang Wang, Chenjing Ye, Junmin Li, Zhen Jin, Rufang Xiang","doi":"10.3724/abbs.2025156","DOIUrl":"https://doi.org/10.3724/abbs.2025156","url":null,"abstract":"<p><p>CD19-directed chimeric antigen receptor T (CAR-T) cell therapy is promising for treating relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL), yet its long-term efficacy remains limited by CAR-T-cell exhaustion. Given the broad immunomodulatory activities of vitamin D, we investigate whether its active form, 1α,25(OH) <sub>2</sub>D <sub>3</sub>, enhances CAR-T-cell functionality and improves therapeutic outcomes. We demonstrate that 1α,25(OH) <sub>2</sub>D <sub>3</sub> treatment significantly mitigates exhaustion and enhances the antitumor activity of CD19 CAR-T cells derived from both healthy donors and DLBCL patients, which is further validated in xenograft mouse models. Mechanistically, we show that 1α,25(OH) <sub>2</sub>D <sub>3</sub> upregulates the expression of the vitamin D receptor (VDR), promoting transcriptional reprogramming associated with memory-like differentiation and downregulation of exhaustion-related genes, thereby reshaping the functional heterogeneity of CAR-T cells under tumor stimulation. Our study highlights 1α,25(OH) <sub>2</sub>D <sub>3</sub> supplementation as a safe and accessible approach to mitigate terminal differentiation and exhaustion of CAR-T cells, offering a promising strategy to enhance the clinical efficacy of CAR-T therapy in patients with R/R DLBCL.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720251","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}
CD47, a cell surface transmembrane glycoprotein, is an innate immune checkpoint that suppresses phagocytic clearance. Emerging evidence suggests that CD47 has noncanonical functions. However, its involvement in chemotherapy resistance is not well understood. Our study reveals that cisplatin treatment upregulates CD47 expression across multiple cancer cell lines. Cisplatin induces the expression of CD47 through the ATM/NF-κB signaling pathway. Genetic ablation of CD47 dramatically sensitizes cancer cells to cisplatin. Mechanistically, CD47 depletion potentiates cisplatin-induced DNA damage, as demonstrated by elevated γH2AX formation and ATM phosphorylation. Knockdown of CD47 inhibits the expression of DNA repair genes ERCC1, FANCA, and BRCA2 through the ATM/NF-κB pathway. Remarkably, CD47 blockade with neutralizing antibodies recapitulates these effects, synergistically potentiating cisplatin's DNA-damaging capacity while suppressing DNA repair capacity. CD47 blockade also potentiates cisplatin's tumor inhibitory effect in vivo. These findings establish a novel mechanism whereby CD47 promotes cisplatin resistance through transcriptional regulation of DNA repair pathway, providing rationale for combining CD47-targeted therapies with conventional chemotherapy. This dual approach could simultaneously overcome immune evasion while enhancing treatment efficacy.
{"title":"CD47 blockade enhances cisplatin sensitivity by inhibiting DNA repair gene expression.","authors":"Xingqian Liu, Jie Lun, Jianxin Xu, Liyuan Jing, Yuying Zhang, Yu Wang, Zhengyu Jin, Mengchao Yu, Jing Fang","doi":"10.3724/abbs.2025147","DOIUrl":"https://doi.org/10.3724/abbs.2025147","url":null,"abstract":"<p><p>CD47, a cell surface transmembrane glycoprotein, is an innate immune checkpoint that suppresses phagocytic clearance. Emerging evidence suggests that CD47 has noncanonical functions. However, its involvement in chemotherapy resistance is not well understood. Our study reveals that cisplatin treatment upregulates CD47 expression across multiple cancer cell lines. Cisplatin induces the expression of CD47 through the ATM/NF-κB signaling pathway. Genetic ablation of CD47 dramatically sensitizes cancer cells to cisplatin. Mechanistically, CD47 depletion potentiates cisplatin-induced DNA damage, as demonstrated by elevated γH2AX formation and ATM phosphorylation. Knockdown of <i>CD47</i> inhibits the expression of DNA repair genes <i>ERCC1</i>, <i>FANCA</i>, and <i>BRCA2</i> through the ATM/NF-κB pathway. Remarkably, CD47 blockade with neutralizing antibodies recapitulates these effects, synergistically potentiating cisplatin's DNA-damaging capacity while suppressing DNA repair capacity. CD47 blockade also potentiates cisplatin's tumor inhibitory effect <i>in vivo</i>. These findings establish a novel mechanism whereby CD47 promotes cisplatin resistance through transcriptional regulation of DNA repair pathway, providing rationale for combining CD47-targeted therapies with conventional chemotherapy. This dual approach could simultaneously overcome immune evasion while enhancing treatment efficacy.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145720289","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}
Hanfei Gao, Chaomei Li, Jie Sun, Liyuan Deng, Jia Li, Zhiqiang Wu, Hu Chen
Lung squamous cell carcinoma (LUSC) remains a major therapeutic challenge because of its pronounced resistance to chemotherapy, particularly carboplatin. In this study, we investigate the role of SOX2, a lineage-survival oncogene, in mediating carboplatin resistance in LUSC. We demonstrate that SOX2 is highly expressed in LUSC and is significantly associated with poor prognosis. Our results show that SOX2 directly transactivates the expression of NRF2, a master regulator of cellular redox homeostasis, thereby increasing glutathione (GSH) synthesis and protecting cells from carboplatin-induced oxidative stress. Pharmacological or genetic inhibition of NRF2 effectively abrogates SOX2-mediated carboplatin resistance both in vitro and in vivo, resensitizing LUSC cells to chemotherapy. These findings highlight SOX2 as a critical redox regulator that modulates NRF2 signaling to promote carboplatin resistance in LUSC. The identification of the SOX2-NRF2 axis as a potential therapeutic target suggests that NRF2 inhibition may represent a promising strategy to overcome chemoresistance in LUSC.
{"title":"SOX2 transactivates NRF2 to promote carboplatin resistance in lung squamous cell carcinoma.","authors":"Hanfei Gao, Chaomei Li, Jie Sun, Liyuan Deng, Jia Li, Zhiqiang Wu, Hu Chen","doi":"10.3724/abbs.2025228","DOIUrl":"https://doi.org/10.3724/abbs.2025228","url":null,"abstract":"<p><p>Lung squamous cell carcinoma (LUSC) remains a major therapeutic challenge because of its pronounced resistance to chemotherapy, particularly carboplatin. In this study, we investigate the role of SOX2, a lineage-survival oncogene, in mediating carboplatin resistance in LUSC. We demonstrate that SOX2 is highly expressed in LUSC and is significantly associated with poor prognosis. Our results show that SOX2 directly transactivates the expression of NRF2, a master regulator of cellular redox homeostasis, thereby increasing glutathione (GSH) synthesis and protecting cells from carboplatin-induced oxidative stress. Pharmacological or genetic inhibition of NRF2 effectively abrogates SOX2-mediated carboplatin resistance both <i>in vitro</i> and <i>in vivo</i>, resensitizing LUSC cells to chemotherapy. These findings highlight SOX2 as a critical redox regulator that modulates NRF2 signaling to promote carboplatin resistance in LUSC. The identification of the SOX2-NRF2 axis as a potential therapeutic target suggests that NRF2 inhibition may represent a promising strategy to overcome chemoresistance in LUSC.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145686791","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}
Wenjian Wang, Jinglu Yu, Tanxuan Huang, Kangdi Liu, Lianxiang Luo
Ferroptosis, an iron-dependent form of regulated cell death, is characterized by excessive reactive oxygen species (ROS) accumulation and lipid peroxidation of polyunsaturated fatty acids (PUFAs) in cellular membranes. Non-small cell lung cancer (NSCLC) harboring KRAS mutations often exhibits therapeutic resistance but may display high susceptibility to ferroptosis. Eupalinolide B (EB), a natural compound with documented anti-cancer activity, has not been thoroughly explored for its ferroptosis-inducing potential in KRAS-mutant NSCLC. In this study, we demonstrate that EB treatment significantly elevates ROS levels, intracellular iron accumulation, and lipid peroxidation in KRAS-mutant NSCLC cells, resulting in ferroptotic cell death. Molecular docking and cellular thermal shift assays reveal that EB directly binds to and activates heme oxygenase-1 (HO-1), a critical component of the Kelch-like ECH-associated protein 1 (Keap1)-Nrf2/HO-1 oxidative stress response pathway. Genetic or pharmacological inhibition of HO-1 attenuates EB-induced ferroptosis, underscoring the pivotal role of HO-1-mediated oxidative stress in this process. Furthermore, in vivo studies using KRAS-mutant H358 xenograft models confirm the potent anti-tumor effects of EB. Collectively, our findings establish that EB triggers ferroptosis in KRAS-mutant NSCLC by activating the Keap1-Nrf2/HO-1 pathway, suggesting a promising therapeutic strategy for this challenging malignancy.
铁死亡是一种铁依赖性的细胞死亡形式,其特征是细胞膜中活性氧(ROS)的过度积累和多不饱和脂肪酸(PUFAs)的脂质过氧化。携带KRAS突变的非小细胞肺癌(NSCLC)通常表现出治疗耐药性,但可能对铁下垂表现出高易感性。Eupalinolide B (EB)是一种具有抗癌活性的天然化合物,但其在kras突变型NSCLC中诱导铁凋亡的潜力尚未得到充分的研究。在这项研究中,我们证明EB治疗显著提高kras突变的NSCLC细胞中的ROS水平、细胞内铁积累和脂质过氧化,导致铁致细胞死亡。分子对接和细胞热移实验表明,EB直接结合并激活血红素加氧酶-1 (HO-1),这是kelch样ech相关蛋白1 (Keap1)-Nrf2/HO-1氧化应激反应途径的关键组成部分。遗传或药理抑制HO-1可减轻eb诱导的铁下垂,强调HO-1介导的氧化应激在这一过程中的关键作用。此外,利用kras突变体H358异种移植模型进行的体内研究证实了EB的有效抗肿瘤作用。总的来说,我们的研究结果表明,EB通过激活Keap1-Nrf2/HO-1通路,在kras突变的NSCLC中触发铁凋亡,这为这种具有挑战性的恶性肿瘤提供了一种有希望的治疗策略。
{"title":"Eupalinolide B exerts cytotoxic effects against KRAS-mutant NSCLC through Nrf2/HO-1-regulated ferroptosis.","authors":"Wenjian Wang, Jinglu Yu, Tanxuan Huang, Kangdi Liu, Lianxiang Luo","doi":"10.3724/abbs.2025211","DOIUrl":"https://doi.org/10.3724/abbs.2025211","url":null,"abstract":"<p><p>Ferroptosis, an iron-dependent form of regulated cell death, is characterized by excessive reactive oxygen species (ROS) accumulation and lipid peroxidation of polyunsaturated fatty acids (PUFAs) in cellular membranes. Non-small cell lung cancer (NSCLC) harboring KRAS mutations often exhibits therapeutic resistance but may display high susceptibility to ferroptosis. Eupalinolide B (EB), a natural compound with documented anti-cancer activity, has not been thoroughly explored for its ferroptosis-inducing potential in KRAS-mutant NSCLC. In this study, we demonstrate that EB treatment significantly elevates ROS levels, intracellular iron accumulation, and lipid peroxidation in KRAS-mutant NSCLC cells, resulting in ferroptotic cell death. Molecular docking and cellular thermal shift assays reveal that EB directly binds to and activates heme oxygenase-1 (HO-1), a critical component of the Kelch-like ECH-associated protein 1 (Keap1)-Nrf2/HO-1 oxidative stress response pathway. Genetic or pharmacological inhibition of HO-1 attenuates EB-induced ferroptosis, underscoring the pivotal role of HO-1-mediated oxidative stress in this process. Furthermore, <i>in vivo</i> studies using KRAS-mutant H358 xenograft models confirm the potent anti-tumor effects of EB. Collectively, our findings establish that EB triggers ferroptosis in KRAS-mutant NSCLC by activating the Keap1-Nrf2/HO-1 pathway, suggesting a promising therapeutic strategy for this challenging malignancy.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145675864","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}
Xinyuan Zhu, Hongyan Zhai, Huishuang Shao, Dawei Wu, Jun Ren, Daqing Sun, Sujuan Liu
Sepsis-induced cardiomyopathy (SIC) is a severe complication of sepsis and septic shock and is characterized by cardiac dysfunction. Levosimendan (LEVO), a calcium sensitizer, has shown therapeutic potential in SIC, although its underlying mechanism remains unclear. Nrf2, a pivotal regulator of antioxidant and anti-inflammatory responses, may represent a potential target for SIC treatment. In this study, we examine the effects of LEVO on SIC and explore the mechanistic role of Nrf2 in mediating its cardioprotective effects. A murine SIC model is established via cecal ligation and puncture (CLP), and cardiomyocyte injury is induced in vitro via lipopolysaccharide (LPS) exposure in HL-1 cells. The CLP procedure significantly elevates serum cTnI and IL-6 levels and reduces the survival rates of mice. Echocardiographic analysis reveals impaired cardiac structure and function, accompanied by mitochondrial morphological and functional damage, in SIC mice. Interestingly, these pathological changes in SIC are markedly attenuated by LEVO treatment. Similarly, LEVO administration restores proliferative capacity; increases mitochondrial ATP, mitochondrial membrane potential (MMP) and NADH levels; and reduces ROS production and intracellular calcium overload. Notably, the protective effects of LEVO on cardiomyocyte viability and mitochondrial function are significantly diminished following Nrf2 inhibition or Nrf2 knockout (KO). Collectively, these findings demonstrate that LEVO mitigates cardiomyocyte injury and mitochondrial dysfunction in SIC through an Nrf2-dependent mechanism.
{"title":"Levosimendan ameliorates cardiomyocyte injury and mitochondrial dysfunction in an Nrf2-dependent manner in mice with sepsis-induced cardiomyopathy.","authors":"Xinyuan Zhu, Hongyan Zhai, Huishuang Shao, Dawei Wu, Jun Ren, Daqing Sun, Sujuan Liu","doi":"10.3724/abbs.2025165","DOIUrl":"https://doi.org/10.3724/abbs.2025165","url":null,"abstract":"<p><p>Sepsis-induced cardiomyopathy (SIC) is a severe complication of sepsis and septic shock and is characterized by cardiac dysfunction. Levosimendan (LEVO), a calcium sensitizer, has shown therapeutic potential in SIC, although its underlying mechanism remains unclear. Nrf2, a pivotal regulator of antioxidant and anti-inflammatory responses, may represent a potential target for SIC treatment. In this study, we examine the effects of LEVO on SIC and explore the mechanistic role of Nrf2 in mediating its cardioprotective effects. A murine SIC model is established via cecal ligation and puncture (CLP), and cardiomyocyte injury is induced <i>in vitro</i> via lipopolysaccharide (LPS) exposure in HL-1 cells. The CLP procedure significantly elevates serum cTnI and IL-6 levels and reduces the survival rates of mice. Echocardiographic analysis reveals impaired cardiac structure and function, accompanied by mitochondrial morphological and functional damage, in SIC mice. Interestingly, these pathological changes in SIC are markedly attenuated by LEVO treatment. Similarly, LEVO administration restores proliferative capacity; increases mitochondrial ATP, mitochondrial membrane potential (MMP) and NADH levels; and reduces ROS production and intracellular calcium overload. Notably, the protective effects of LEVO on cardiomyocyte viability and mitochondrial function are significantly diminished following Nrf2 inhibition or <i>Nrf2</i> knockout (KO). Collectively, these findings demonstrate that LEVO mitigates cardiomyocyte injury and mitochondrial dysfunction in SIC through an Nrf2-dependent mechanism.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145666756","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}
Benrong Liu, Lei Fang, Chunxia Miao, Xinyu Wen, Xiumiao Zheng, Minxing Xu, Junli Lin, Yujuan Xiong, Shi-Ming Liu
Myocyte enhancer factor 2A (MEF2A), a transcription factor implicated in coronary artery disease, remains unexplored in vascular redox regulation. To address this gap and overcome the limitations of current antioxidant therapies, we investigate the role of MEF2A in oxidative defense via human umbilical vein endothelial cells (HUVECs) and murine models. Adenoviral vectors encoding MEF2A-specific shRNAs or mRNAs are used to silence or overexpress MEF2A in HUVECs. For in vivo validation, endothelial-targeted MEF2A knockdown is achieved via AAV1-shRNA delivery in mice fed with a high-fat diet. Systemic redox status is assessed by measuring reactive oxygen species (ROS), glutathione homeostasis (GSH/GSSG ratio), the NADH/NAD + balance, the mitochondrial membrane potential (ΔΨm), and 8-hydroxy-2'-deoxyguanosine (8-OHdG). Mechanistic insights are derived from immunofluorescence, qPCR, western blotting, and dual-luciferase reporter assays. MEF2A silencing induces redox imbalance, characterized by elevated ROS, a reduced GSH/GSSG ratio, and ΔΨm collapse. Conversely, MEF2A overexpression synergizes with SIRT1 to restore the glutathione pool, maintain NAD + homeostasis, and suppress ROS under oxidative stress. Chromatin immunoprecipitation confirms that MEF2A directly binds to two cis-elements in the SIRT1 promoter, driving transcriptional activation. In vivo, MEF2A-deficient mice present increased vascular oxidative damage, as indicated by elevated DNA damage marker (8-OHdG) and ROS levels. The downregulation of SIRT1/PGC-1α in MEF2A-silenced cells is verified in vivo. Our findings establish MEF2A as a master regulator of endothelial redox defense via the SIRT1-PGC-1α axis, providing a mechanistic foundation for the treatment of oxidative cardiovascular disorders. This work suggests that pharmacological MEF2A activation is a novel strategy for precision antioxidant therapy in vascular medicine.
肌细胞增强因子2A (MEF2A)是一种与冠状动脉疾病有关的转录因子,在血管氧化还原调节中仍未被发现。为了解决这一空白并克服当前抗氧化疗法的局限性,我们通过人脐静脉内皮细胞(HUVECs)和小鼠模型研究了MEF2A在氧化防御中的作用。在HUVECs中,利用腺病毒载体编码MEF2A特异性shrna或mrna沉默或过表达MEF2A。为了在体内验证,在喂食高脂肪饮食的小鼠中,通过递送AAV1-shRNA实现了内皮靶向MEF2A的敲除。通过测定活性氧(ROS)、谷胱甘肽稳态(GSH/GSSG比值)、NADH/NAD +平衡、线粒体膜电位(ΔΨm)和8-羟基-2′-脱氧鸟苷(8-OHdG)来评估全身氧化还原状态。机制见解来自免疫荧光,qPCR, western blotting和双荧光素酶报告分析。MEF2A沉默诱导氧化还原失衡,其特征是ROS升高、GSH/GSSG比例降低和ΔΨm崩溃。相反,MEF2A过表达与SIRT1协同恢复谷胱甘肽库,维持NAD +稳态,抑制氧化应激下的ROS。染色质免疫沉淀证实,MEF2A直接结合SIRT1启动子中的两个顺式元件,驱动转录激活。在体内,mef2a缺陷小鼠血管氧化损伤增加,DNA损伤标志物(8-OHdG)和ROS水平升高。体内实验证实了mef2a沉默细胞SIRT1/PGC-1α的下调。我们的研究结果证实MEF2A通过SIRT1-PGC-1α轴调控内皮氧化还原防御,为氧化性心血管疾病的治疗提供了机制基础。这项研究表明,MEF2A的药理激活是血管医学中精确抗氧化治疗的一种新策略。
{"title":"Myocyte enhancer factor 2a orchestrates vascular redox homeostasis via direct transcriptional activation of SIRT1.","authors":"Benrong Liu, Lei Fang, Chunxia Miao, Xinyu Wen, Xiumiao Zheng, Minxing Xu, Junli Lin, Yujuan Xiong, Shi-Ming Liu","doi":"10.3724/abbs.2025163","DOIUrl":"https://doi.org/10.3724/abbs.2025163","url":null,"abstract":"<p><p>Myocyte enhancer factor 2A (MEF2A), a transcription factor implicated in coronary artery disease, remains unexplored in vascular redox regulation. To address this gap and overcome the limitations of current antioxidant therapies, we investigate the role of MEF2A in oxidative defense via human umbilical vein endothelial cells (HUVECs) and murine models. Adenoviral vectors encoding MEF2A-specific shRNAs or mRNAs are used to silence or overexpress <i>MEF2A</i> in HUVECs. For <i>in vivo</i> validation, endothelial-targeted <i>MEF2A</i> knockdown is achieved via AAV1-shRNA delivery in mice fed with a high-fat diet. Systemic redox status is assessed by measuring reactive oxygen species (ROS), glutathione homeostasis (GSH/GSSG ratio), the NADH/NAD <sup>+</sup> balance, the mitochondrial membrane potential (ΔΨm), and 8-hydroxy-2'-deoxyguanosine (8-OHdG). Mechanistic insights are derived from immunofluorescence, qPCR, western blotting, and dual-luciferase reporter assays. <i>MEF2A</i> silencing induces redox imbalance, characterized by elevated ROS, a reduced GSH/GSSG ratio, and ΔΨm collapse. Conversely, MEF2A overexpression synergizes with SIRT1 to restore the glutathione pool, maintain NAD <sup>+</sup> homeostasis, and suppress ROS under oxidative stress. Chromatin immunoprecipitation confirms that MEF2A directly binds to two cis-elements in the <i>SIRT1</i> promoter, driving transcriptional activation. <i>In vivo</i>, MEF2A-deficient mice present increased vascular oxidative damage, as indicated by elevated DNA damage marker (8-OHdG) and ROS levels. The downregulation of SIRT1/PGC-1α in <i>MEF2A</i>-silenced cells is verified <i>in vivo</i>. Our findings establish MEF2A as a master regulator of endothelial redox defense via the SIRT1-PGC-1α axis, providing a mechanistic foundation for the treatment of oxidative cardiovascular disorders. This work suggests that pharmacological MEF2A activation is a novel strategy for precision antioxidant therapy in vascular medicine.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-12-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145666761","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}
Glycolysis provides the main energy source for the rapid proliferation and migration of colorectal cancer (CRC) cells. In our previous studies, we reported that SUN5, a nuclear membrane protein, promotes proliferation and migration. However, whether SUN5 is involved in the process of glycolysis is unclear. Here, we demonstrate that overexpression of SUN5 enhances glucose uptake and lactate production in CRC cells, whereas the opposite results are observed in SUN5-knockdown cells. Mechanistically, SUN5 activates the NF-κB signaling pathway, which can be inhibited by the IKK inhibitor BAY11-7082. Further studies reveal that SUN5 interacts with TRIM28 to increase IκB α ubiquitination, leading to the nuclear translocation of phosphorylated P65 (phos-P65) and subsequent increases in the transcription of GLUT1 and LDHA, accelerating glycolysis. Moreover, xenograft transplantation experiments reveal that the knockdown of SUN5 inhibits glycolysis and tumorigenesis in vivo. Taken together, these findings indicate that SUN5 enhances the glycolysis and tumorigenesis of CRC cells via interaction with TRIM28, which provides a potential target for the diagnosis and treatment of CRC.
{"title":"SUN5 interacts with TRIM28, enhancing IκBα ubiquitination to promote glycolysis in colorectal cancer cells.","authors":"Jingyuan Chen, Youbo Yang, Gang Liu, Lihua Huang, Yunfei Zhang, Yufeng Wang, Xiuwen Xu, Xiaowei Xing","doi":"10.3724/abbs.2025201","DOIUrl":"https://doi.org/10.3724/abbs.2025201","url":null,"abstract":"<p><p>Glycolysis provides the main energy source for the rapid proliferation and migration of colorectal cancer (CRC) cells. In our previous studies, we reported that SUN5, a nuclear membrane protein, promotes proliferation and migration. However, whether SUN5 is involved in the process of glycolysis is unclear. Here, we demonstrate that overexpression of SUN5 enhances glucose uptake and lactate production in CRC cells, whereas the opposite results are observed in <i>SUN5</i>-knockdown cells. Mechanistically, SUN5 activates the NF-κB signaling pathway, which can be inhibited by the IKK inhibitor BAY11-7082. Further studies reveal that SUN5 interacts with TRIM28 to increase IκB <i>α</i> ubiquitination, leading to the nuclear translocation of phosphorylated P65 (phos-P65) and subsequent increases in the transcription of <i>GLUT1</i> and <i>LDHA</i>, accelerating glycolysis. Moreover, xenograft transplantation experiments reveal that the knockdown of <i>SUN5</i> inhibits glycolysis and tumorigenesis <i>in vivo</i>. Taken together, these findings indicate that SUN5 enhances the glycolysis and tumorigenesis of CRC cells via interaction with TRIM28, which provides a potential target for the diagnosis and treatment of CRC.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145627770","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}
Yinghui Qiu, Chunlan Li, Peiying Ye, Haiyun Zhang, Yanxiu Liu, Weiyan Ma, Chen Lin, Rongqin Ke
Spatial biology aims to elucidate cellular organization, function, and interactions within native tissue contexts, offering key insights into both normal physiology and disease. Spatial proteomics complements this by enabling high-resolution mapping of protein localization and abundance, directly reflecting functional cellular states. Unlike transcriptomics, which infers potential activity, proteomics captures actual molecular functions, including post-translational modifications and dynamic interactions. However, in situ protein profiling poses significant challenges, as proteins cannot be directly sequenced or easily targeted via nucleic acid hybridization. Antibody-oligonucleotide conjugates (AOCs) address this limitation by converting protein recognition into a DNA-based readout, thereby enabling sensitive and scalable detection. In this review, we outline the core principles of AOC-based spatial proteomic technologies, including multiplexed protein analysis, in situ protein-protein interactions, and integration with other biomolecular data. We highlight their applications in decoding tissue complexity and disease pathology and examine key technical challenges that remain. Overall, AOCs offer distinct advantages, including DNA-mediated signal amplification, spatially resolved proteomic profiling, and compatibility with multi-omics approaches, positioning them as powerful platforms in the advancement of spatial biology.
{"title":"Antibody-oligonucleotide conjugates for spatial proteomics: principles, applications, and challenges.","authors":"Yinghui Qiu, Chunlan Li, Peiying Ye, Haiyun Zhang, Yanxiu Liu, Weiyan Ma, Chen Lin, Rongqin Ke","doi":"10.3724/abbs.2025212","DOIUrl":"https://doi.org/10.3724/abbs.2025212","url":null,"abstract":"<p><p>Spatial biology aims to elucidate cellular organization, function, and interactions within native tissue contexts, offering key insights into both normal physiology and disease. Spatial proteomics complements this by enabling high-resolution mapping of protein localization and abundance, directly reflecting functional cellular states. Unlike transcriptomics, which infers potential activity, proteomics captures actual molecular functions, including post-translational modifications and dynamic interactions. However, <i>in situ</i> protein profiling poses significant challenges, as proteins cannot be directly sequenced or easily targeted via nucleic acid hybridization. Antibody-oligonucleotide conjugates (AOCs) address this limitation by converting protein recognition into a DNA-based readout, thereby enabling sensitive and scalable detection. In this review, we outline the core principles of AOC-based spatial proteomic technologies, including multiplexed protein analysis, <i>in situ</i> protein-protein interactions, and integration with other biomolecular data. We highlight their applications in decoding tissue complexity and disease pathology and examine key technical challenges that remain. Overall, AOCs offer distinct advantages, including DNA-mediated signal amplification, spatially resolved proteomic profiling, and compatibility with multi-omics approaches, positioning them as powerful platforms in the advancement of spatial biology.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145627702","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}
Emerging studies have revealed that disruptions in circadian crosstalk between the gut microbiota and the host play an essential role in the pathogenesis of metabolic disorders. Under physiological conditions, host circadian clocks regulate microbial diurnal oscillations through rhythmic behaviors, including feeding patterns and sleep-wake cycles. This temporal regulation manifests as robust 24-hour oscillations in microbial community composition, spatial organization, and metabolic activity. These rhythmic microbial signals and their metabolic outputs are subsequently translated into host immune modulation, establishing a bidirectional temporal dialogue between the host and microbiota. Modern lifestyle disruptions, including erratic eating patterns and shift work, desynchronize this temporal dialogue, leading to the loss of microbial rhythms, impaired intestinal barrier function, maladaptive immune responses, chronic inflammation, and systemic metabolic dysregulation. This review delineates the mechanisms through which host-microbiota circadian crosstalk governs immunometabolic homeostasis, provides a mechanistic framework for understanding immunometabolic diseases, and highlights therapeutic strategies that target microbial rhythms to reset host immunity and metabolism.
{"title":"Gut microbiota circadian rhythms: a key regulator of immunometabolic homeostasis.","authors":"Zitong Zhao, Siyan Wu, Tingting Wang, Yue Zhao","doi":"10.3724/abbs.2025220","DOIUrl":"https://doi.org/10.3724/abbs.2025220","url":null,"abstract":"<p><p>Emerging studies have revealed that disruptions in circadian crosstalk between the gut microbiota and the host play an essential role in the pathogenesis of metabolic disorders. Under physiological conditions, host circadian clocks regulate microbial diurnal oscillations through rhythmic behaviors, including feeding patterns and sleep-wake cycles. This temporal regulation manifests as robust 24-hour oscillations in microbial community composition, spatial organization, and metabolic activity. These rhythmic microbial signals and their metabolic outputs are subsequently translated into host immune modulation, establishing a bidirectional temporal dialogue between the host and microbiota. Modern lifestyle disruptions, including erratic eating patterns and shift work, desynchronize this temporal dialogue, leading to the loss of microbial rhythms, impaired intestinal barrier function, maladaptive immune responses, chronic inflammation, and systemic metabolic dysregulation. This review delineates the mechanisms through which host-microbiota circadian crosstalk governs immunometabolic homeostasis, provides a mechanistic framework for understanding immunometabolic diseases, and highlights therapeutic strategies that target microbial rhythms to reset host immunity and metabolism.</p>","PeriodicalId":6978,"journal":{"name":"Acta biochimica et biophysica Sinica","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145627707","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}