Pub Date : 2025-03-08DOI: 10.1007/s10495-025-02078-0
Jialin Zheng, Xiaoyu Xu, Ziwei Zhang, Kanghui Ge, Yi Xiang, Hualei Dai
Object
Acute myocardial infarction (AMI) is a serious cardiovascular disease for which there are still no effective therapeutic options available, and melanoma-associated antigen-A13 (Magea13), a member of the MAGE superfamily, has an unknown role in AMI. This study aims to investigate the potential role and molecular mechanisms of Magea13 in myocardial injury associated with AMI through in vivo and in vitro experiments.
Methods
Firstly, differentially expressed genes (DEGs) and signaling pathways were screened by RNA sequencing. Cardiac-specific Magea13 overexpression was achieved with the adeno-associated virus type 9 serotype system. Subsequently, these rats underwent left anterior descending coronary artery (LAD) ligation, followed by histopathological examination, biochemical assay, and Western blot analysis to evaluate the efficacy and feasibility of Magea13 in AMI. Meanwhile, the Magea13-overexpressing rat cardiomyocyte cell line (H9c2) was also subjected to hypoxia-glucose deficiency/reperfusion to mimic AMI injury to further validate its effects in vitro.
Results
The cardiomyocyte-specific overexpression of Magea13 was observed to attenuate myocardial injury in rats with acute myocardial infarction. Furthermore, Magea13 overexpression was demonstrated to attenuate OGD/R-induced H9c2 cell injury. Mechanistic studies have suggested that the protective effect of Magea13 may be mediated through the cAMP-PKA pathway.
Conclusion
Magea13 has been demonstrated to offer protection against AMI myocardial injury through the cAMP-PKA signaling pathway and is therefore a promising therapeutic and predictive target for AMI myocardial injury.
{"title":"Magea13 attenuates myocardial injury in acute myocardial infarction by inhibiting the cAMP-PKA signaling pathway","authors":"Jialin Zheng, Xiaoyu Xu, Ziwei Zhang, Kanghui Ge, Yi Xiang, Hualei Dai","doi":"10.1007/s10495-025-02078-0","DOIUrl":"10.1007/s10495-025-02078-0","url":null,"abstract":"<div><h3>Object</h3><p>Acute myocardial infarction (AMI) is a serious cardiovascular disease for which there are still no effective therapeutic options available, and melanoma-associated antigen-A13 (Magea13), a member of the MAGE superfamily, has an unknown role in AMI. This study aims to investigate the potential role and molecular mechanisms of Magea13 in myocardial injury associated with AMI through in vivo and in vitro experiments.</p><h3>Methods</h3><p>Firstly, differentially expressed genes (DEGs) and signaling pathways were screened by RNA sequencing. Cardiac-specific Magea13 overexpression was achieved with the adeno-associated virus type 9 serotype system. Subsequently, these rats underwent left anterior descending coronary artery (LAD) ligation, followed by histopathological examination, biochemical assay, and Western blot analysis to evaluate the efficacy and feasibility of Magea13 in AMI. Meanwhile, the Magea13-overexpressing rat cardiomyocyte cell line (H9c2) was also subjected to hypoxia-glucose deficiency/reperfusion to mimic AMI injury to further validate its effects in vitro.</p><h3>Results</h3><p>The cardiomyocyte-specific overexpression of Magea13 was observed to attenuate myocardial injury in rats with acute myocardial infarction. Furthermore, Magea13 overexpression was demonstrated to attenuate OGD/R-induced H9c2 cell injury. Mechanistic studies have suggested that the protective effect of Magea13 may be mediated through the cAMP-PKA pathway.</p><h3>Conclusion</h3><p>Magea13 has been demonstrated to offer protection against AMI myocardial injury through the cAMP-PKA signaling pathway and is therefore a promising therapeutic and predictive target for AMI myocardial injury.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"30 3-4","pages":"1042 - 1057"},"PeriodicalIF":6.1,"publicationDate":"2025-03-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s10495-025-02078-0.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143582165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Diabetes mellitus is a common chronic metabolic disease, with its prevalence escalating annually. Diabetic cardiomyopathy is a leading cause of mortality among diabetic patients, characterized by intricate metabolic disturbances and myocardial cell demise. Various forms of cellular death pathways including apoptosis, pyroptosis, autophagic cell death, necroptosis, ferroptosis, and entosis have been identified in diabetic cardiomyopathy. Inhibiting myocardial cell death pathways has shown promise in mitigating diabetic cardiomyopathy progression. However, there are still gaps in understanding the role of metal ions in diabetic cardiomyopathy pathogenesis. Recent research endeavors have found that iron, copper, zinc, calcium, manganese and other metal elements related to cell death play an intricate and critical role in the pathogenesis and progression of diabetic cardiomyopathy. Notably, many animal studies have shown that the development and progression of diabetic cardiomyopathy can be alleviated by inhibiting the cell death process induced by these metal ions. Therefore, we review the molecular mechanisms underlying the death of various metal ions and the potential pathophysiological roles they play in diabetic cardiomyopathy. In addition, the value of these metal ions in the treatment of diabetic cardiomyopathy is also described.
{"title":"Novel impact of metal ion-induced cell death on diabetic cardiomyopathy pathogenesis and therapy.","authors":"Jingjing Jiang, Shengnan Hu, Kaibo Hu, Leyang Xiao, Jitao Lin, Yixuan Chen, Deju Zhang, Yangliu Ou, Jing Zhang, Linhui Yuan, Wenting Wang, Peng Yu","doi":"10.1007/s10495-025-02090-4","DOIUrl":"https://doi.org/10.1007/s10495-025-02090-4","url":null,"abstract":"<p><p>Diabetes mellitus is a common chronic metabolic disease, with its prevalence escalating annually. Diabetic cardiomyopathy is a leading cause of mortality among diabetic patients, characterized by intricate metabolic disturbances and myocardial cell demise. Various forms of cellular death pathways including apoptosis, pyroptosis, autophagic cell death, necroptosis, ferroptosis, and entosis have been identified in diabetic cardiomyopathy. Inhibiting myocardial cell death pathways has shown promise in mitigating diabetic cardiomyopathy progression. However, there are still gaps in understanding the role of metal ions in diabetic cardiomyopathy pathogenesis. Recent research endeavors have found that iron, copper, zinc, calcium, manganese and other metal elements related to cell death play an intricate and critical role in the pathogenesis and progression of diabetic cardiomyopathy. Notably, many animal studies have shown that the development and progression of diabetic cardiomyopathy can be alleviated by inhibiting the cell death process induced by these metal ions. Therefore, we review the molecular mechanisms underlying the death of various metal ions and the potential pathophysiological roles they play in diabetic cardiomyopathy. In addition, the value of these metal ions in the treatment of diabetic cardiomyopathy is also described.</p>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143555659","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}
Pub Date : 2025-03-05DOI: 10.1007/s10495-025-02098-w
Jianxin Cheng, Jin Gao, Jianjun Li, Hao Tian
Postoperative cognitive dysfunction (POCD) is a prevalent complication affecting the central nervous system after surgery, manifesting as a decline in cognitive abilities, particularly common among elderly patients. Surgical stress and anesthesia can activate systemic inflammation, prompting immune cells, including neutrophils, to infiltrate the brain, thereby triggering neuroinflammation and resulting in cognitive impairment. Neutrophils, as crucial effector cells in innate immunity, have been increasingly recognized in recent years for their significant role in the pathogenesis of POCD due to their vital function in inflammatory responses. They are not only rapidly activated in peripheral blood, secreting a range of cytokines, chemokines, and neutrophil extracellular traps (NETs), but also possess the capacity to alter the permeability of the blood-brain barrier (BBB), further facilitating the development of neuroinflammation. This paper systematically reviews the recent findings on the diverse functions of neutrophils and their role in POCD, aiming to provide novel theoretical foundations and cutting-edge perspectives for advancing foundational research and optimizing clinical intervention strategies for POCD.
{"title":"Neutrophils: a new target for postoperative cognitive dysfunction.","authors":"Jianxin Cheng, Jin Gao, Jianjun Li, Hao Tian","doi":"10.1007/s10495-025-02098-w","DOIUrl":"https://doi.org/10.1007/s10495-025-02098-w","url":null,"abstract":"<p><p>Postoperative cognitive dysfunction (POCD) is a prevalent complication affecting the central nervous system after surgery, manifesting as a decline in cognitive abilities, particularly common among elderly patients. Surgical stress and anesthesia can activate systemic inflammation, prompting immune cells, including neutrophils, to infiltrate the brain, thereby triggering neuroinflammation and resulting in cognitive impairment. Neutrophils, as crucial effector cells in innate immunity, have been increasingly recognized in recent years for their significant role in the pathogenesis of POCD due to their vital function in inflammatory responses. They are not only rapidly activated in peripheral blood, secreting a range of cytokines, chemokines, and neutrophil extracellular traps (NETs), but also possess the capacity to alter the permeability of the blood-brain barrier (BBB), further facilitating the development of neuroinflammation. This paper systematically reviews the recent findings on the diverse functions of neutrophils and their role in POCD, aiming to provide novel theoretical foundations and cutting-edge perspectives for advancing foundational research and optimizing clinical intervention strategies for POCD.</p>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143555658","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}
Autophagy is a vital pathway for recycling and degrading intracellular materials, closely linked to tumorigenesis and progression. The ubiquitin-specific protease (USP) family, as a critical group of deubiquitinating enzymes, plays a complex part in regulating autophagy, metabolism, immune responses, and tumor cells' resistance to drugs. By modifying autophagy-associated proteins through deubiquitination, the USP family influences tumor cell proliferation, survival, and metabolism. Additionally, these enzymes are involved in modulating immune responses within the tumor microenvironment, thereby impacting tumor immune escape. Regarding drug resistance, the USP family enhances the tolerance of tumor cells to chemotherapeutic agents by promoting autophagy. Therefore, targeting USP family members and their regulated autophagy processes may offer new avenues for cancer therapy. This review examines the function of the USP family in tumor autophagy regulation and its implications for tumor progression. The goal of future studies should be to clarify the molecular mechanisms underlying USP-autophagy interactions and their specific roles in various tumor types to establish a theoretical framework for developing novel cancer therapeutic strategies.
{"title":"Role of the USP family in autophagy regulation and cancer progression.","authors":"Congcong Liu, Yalin Yuan, Yuxin Zhan, Mi Zou, Linqian Wu, Chunfang Zhang, Bofan Chen, Haimin Zeng, Ruhui Yang, Tianheng Hu, Jie Peng, Liang Hao","doi":"10.1007/s10495-025-02095-z","DOIUrl":"https://doi.org/10.1007/s10495-025-02095-z","url":null,"abstract":"<p><p>Autophagy is a vital pathway for recycling and degrading intracellular materials, closely linked to tumorigenesis and progression. The ubiquitin-specific protease (USP) family, as a critical group of deubiquitinating enzymes, plays a complex part in regulating autophagy, metabolism, immune responses, and tumor cells' resistance to drugs. By modifying autophagy-associated proteins through deubiquitination, the USP family influences tumor cell proliferation, survival, and metabolism. Additionally, these enzymes are involved in modulating immune responses within the tumor microenvironment, thereby impacting tumor immune escape. Regarding drug resistance, the USP family enhances the tolerance of tumor cells to chemotherapeutic agents by promoting autophagy. Therefore, targeting USP family members and their regulated autophagy processes may offer new avenues for cancer therapy. This review examines the function of the USP family in tumor autophagy regulation and its implications for tumor progression. The goal of future studies should be to clarify the molecular mechanisms underlying USP-autophagy interactions and their specific roles in various tumor types to establish a theoretical framework for developing novel cancer therapeutic strategies.</p>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143555660","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}
Pub Date : 2025-03-05DOI: 10.1007/s10495-025-02092-2
Xiangrong Cui, Huihui Li, Xinyu Zhu, Xia Huang, Tingting Xue, Shu Wang, Xuan Jing
Premature ovarian insufficiency (POI) is a multifactorial condition characterized by diminished ovarian function, granulosa cell (GC) apoptosis, and impaired ovarian angiogenesis, leading to infertility and long-term health complications. Despite its prevalence, effective therapeutic targets for POI remain limited. This study investigates the role of CCDC134 in maintaining ovarian reserve and promoting angiogenesis and its interaction with INHA in a mouse model of POI. Ovarian granulosa cells from POI patients and unaffected women were analyzed for apoptosis and CCDC134 expression. A cisplatin-induced mouse model of POI was used to evaluate the therapeutic potential of AAV-mediated ovary-specific overexpression of CCDC134. Ovarian morphology, hormonal levels, follicular development, granulosa cell viability, and angiogenesis were assessed. The interaction between CCDC134 and INHA was examined using co-immunoprecipitation, immunofluorescence, and molecular pathway analyses. CCDC134 expression was significantly downregulated in ovarian tissues and granulosa cells of POI patients and cisplatin-induced POI mice. CCDC134 overexpression improved ovarian morphology, restored follicular development across all stages, and enhanced reproductive outcomes in POI mice. Hormonal imbalances, including decreased AMH and E2 and elevated FSH and LH, were reversed following CCDC134 overexpression. Moreover, CCDC134 treatment significantly reduced GC apoptosis by downregulating pro-apoptotic markers (Caspase-3 and Bax) and upregulating anti-apoptotic Bcl-2. Angiogenesis was enhanced, as indicated by increased expression of CD34 and vWF, improved endothelial cell viability, and restored VEGF levels. Mechanistic studies revealed a direct interaction between CCDC134 and INHA, with CCDC134 promoting INHA expression and modulating apoptotic and angiogenic pathways. CCDC134 plays a critical role in maintaining ovarian reserve and promoting angiogenesis by directly interacting with INHA. Its overexpression restores ovarian function, mitigates granulosa cell apoptosis, and enhances angiogenesis in a mouse model of POI. These findings highlight the therapeutic potential of the CCDC134-INHA axis as a novel strategy for treating POI.
{"title":"CCDC134 enhances ovarian reserve function and angiogenesis by directly interacting with INHA in a mouse model of premature ovarian insufficiency.","authors":"Xiangrong Cui, Huihui Li, Xinyu Zhu, Xia Huang, Tingting Xue, Shu Wang, Xuan Jing","doi":"10.1007/s10495-025-02092-2","DOIUrl":"https://doi.org/10.1007/s10495-025-02092-2","url":null,"abstract":"<p><p>Premature ovarian insufficiency (POI) is a multifactorial condition characterized by diminished ovarian function, granulosa cell (GC) apoptosis, and impaired ovarian angiogenesis, leading to infertility and long-term health complications. Despite its prevalence, effective therapeutic targets for POI remain limited. This study investigates the role of CCDC134 in maintaining ovarian reserve and promoting angiogenesis and its interaction with INHA in a mouse model of POI. Ovarian granulosa cells from POI patients and unaffected women were analyzed for apoptosis and CCDC134 expression. A cisplatin-induced mouse model of POI was used to evaluate the therapeutic potential of AAV-mediated ovary-specific overexpression of CCDC134. Ovarian morphology, hormonal levels, follicular development, granulosa cell viability, and angiogenesis were assessed. The interaction between CCDC134 and INHA was examined using co-immunoprecipitation, immunofluorescence, and molecular pathway analyses. CCDC134 expression was significantly downregulated in ovarian tissues and granulosa cells of POI patients and cisplatin-induced POI mice. CCDC134 overexpression improved ovarian morphology, restored follicular development across all stages, and enhanced reproductive outcomes in POI mice. Hormonal imbalances, including decreased AMH and E2 and elevated FSH and LH, were reversed following CCDC134 overexpression. Moreover, CCDC134 treatment significantly reduced GC apoptosis by downregulating pro-apoptotic markers (Caspase-3 and Bax) and upregulating anti-apoptotic Bcl-2. Angiogenesis was enhanced, as indicated by increased expression of CD34 and vWF, improved endothelial cell viability, and restored VEGF levels. Mechanistic studies revealed a direct interaction between CCDC134 and INHA, with CCDC134 promoting INHA expression and modulating apoptotic and angiogenic pathways. CCDC134 plays a critical role in maintaining ovarian reserve and promoting angiogenesis by directly interacting with INHA. Its overexpression restores ovarian function, mitigates granulosa cell apoptosis, and enhances angiogenesis in a mouse model of POI. These findings highlight the therapeutic potential of the CCDC134-INHA axis as a novel strategy for treating POI.</p>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143555657","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}
Pub Date : 2025-03-05DOI: 10.1007/s10495-025-02093-1
Hangqi Hu, Jiacheng Zhang, Xiyan Xin, Yuxin Jin, Yutian Zhu, Haolin Zhang, Ruiwen Fan, Yang Ye, Yong Jiang, Dong Li
<p><p>Diminished ovarian reserve (DOR) is a challenging obstacle impacting women' fertility globally with limited treatment option. Bushen Jianpi Tiaoxue Decoction (BJTD) has shown significant efficacy and safety in treating DOR patients, yet the molecular mechanisms behind its effect remain uncertain. Our study aimed to uncover the pharmacology and signaling pathway of BJTD in cyclophosphamide (Cy)-provoked DOR mice and 4-hydroperoxy cyclophosphamide (4-HC)-irritated KGN cells (human granulosa-like cell line) damage models. Granulosa cells from DOR patients and Cy-induced models were reanalyzed utilizing transcriptomics to predict differentially expressed genes and crucial signaling pathways. Validation experiments were conducted in vitro using KGN cells treated with 4-HC, a Cy metabolite, to establish a DOR model. Cell viability, apoptosis, oxidative stress, mitochondrial function, and ferroptosis markers were assessed using the cck8 assay, flow cytometry, fluorescence staining, RT-qPCR, and western blotting analyses. BJTD-serum was evaluated for its protective effects on 4-HC-triggered KGN damages. In vivo, a Cy-induced DOR mouse model was treated with BJTD to evaluate ovarian morphology, estrous cycle, follicle counts, hormone markers, mitochondrial apoptosis and ferroptosis levels, respectively via the vaginal smear, histological analysis, immunostaining, gene and protein expression experiments. The UPLC-MS analysis and network pharmacology were applied to identify BJTD's active ingredients, followed by molecular dockings to assess interactions with the target protein. To confirm the BJTD's mechanism of action, mTOR signaling modulation was analyzed using a specific inhibitor or activator in vitro. Transcriptomic reanalysis revealed significant gene expression differences, with LIF identified as a key target associated with apoptosis pathway. In vitro, 4-HC exposure induced apoptosis, mitochondrial dysfunction, and ferroptosis in KGN cells, accompanied by upregulation of LIF, mTOR, and FoxO3a signalings. BJTD-serum treatment significantly improved cell viability, reduced apoptosis, and alleviated oxidative stress by modulating mitochondrial function and ferroptosis markers, such as Nrf2, HO-1, and GPX4. In vivo, BJTD alleviated Cy-induced ovarian damage, improving ovarian index, estrous cycle, follicle development, and hormone levels, while reducing follicular atresia and granulosa cells apoptosis. Mechanically, BJTD suppressed Cy-induced activation of the LIF-mTOR axis and downstream mitochondrial apoptosis markers, including Cleaved Caspase 9/3, BAX, and γH2AX, while enhancing OPA1 and Bcl-2 expressions. The UPLC-MS outcome combining with network pharmacology identified mainly 20 active compounds in BJTD, with astragaloside IV exhibiting the strongest binding to the mTOR protein. The mTOR pathway modulation experiments confirmed that BJTD's protective effects are mediated through inhibition of hyperactivated mTOR phosphorylation and mitoch
{"title":"Bushen Jianpi Tiaoxue Decoction (BJTD) inhibits the LIF-mTOR signaling axis to regulate mitochondrial function and alleviate cyclophosphamide-induced diminished ovarian reserve.","authors":"Hangqi Hu, Jiacheng Zhang, Xiyan Xin, Yuxin Jin, Yutian Zhu, Haolin Zhang, Ruiwen Fan, Yang Ye, Yong Jiang, Dong Li","doi":"10.1007/s10495-025-02093-1","DOIUrl":"https://doi.org/10.1007/s10495-025-02093-1","url":null,"abstract":"<p><p>Diminished ovarian reserve (DOR) is a challenging obstacle impacting women' fertility globally with limited treatment option. Bushen Jianpi Tiaoxue Decoction (BJTD) has shown significant efficacy and safety in treating DOR patients, yet the molecular mechanisms behind its effect remain uncertain. Our study aimed to uncover the pharmacology and signaling pathway of BJTD in cyclophosphamide (Cy)-provoked DOR mice and 4-hydroperoxy cyclophosphamide (4-HC)-irritated KGN cells (human granulosa-like cell line) damage models. Granulosa cells from DOR patients and Cy-induced models were reanalyzed utilizing transcriptomics to predict differentially expressed genes and crucial signaling pathways. Validation experiments were conducted in vitro using KGN cells treated with 4-HC, a Cy metabolite, to establish a DOR model. Cell viability, apoptosis, oxidative stress, mitochondrial function, and ferroptosis markers were assessed using the cck8 assay, flow cytometry, fluorescence staining, RT-qPCR, and western blotting analyses. BJTD-serum was evaluated for its protective effects on 4-HC-triggered KGN damages. In vivo, a Cy-induced DOR mouse model was treated with BJTD to evaluate ovarian morphology, estrous cycle, follicle counts, hormone markers, mitochondrial apoptosis and ferroptosis levels, respectively via the vaginal smear, histological analysis, immunostaining, gene and protein expression experiments. The UPLC-MS analysis and network pharmacology were applied to identify BJTD's active ingredients, followed by molecular dockings to assess interactions with the target protein. To confirm the BJTD's mechanism of action, mTOR signaling modulation was analyzed using a specific inhibitor or activator in vitro. Transcriptomic reanalysis revealed significant gene expression differences, with LIF identified as a key target associated with apoptosis pathway. In vitro, 4-HC exposure induced apoptosis, mitochondrial dysfunction, and ferroptosis in KGN cells, accompanied by upregulation of LIF, mTOR, and FoxO3a signalings. BJTD-serum treatment significantly improved cell viability, reduced apoptosis, and alleviated oxidative stress by modulating mitochondrial function and ferroptosis markers, such as Nrf2, HO-1, and GPX4. In vivo, BJTD alleviated Cy-induced ovarian damage, improving ovarian index, estrous cycle, follicle development, and hormone levels, while reducing follicular atresia and granulosa cells apoptosis. Mechanically, BJTD suppressed Cy-induced activation of the LIF-mTOR axis and downstream mitochondrial apoptosis markers, including Cleaved Caspase 9/3, BAX, and γH2AX, while enhancing OPA1 and Bcl-2 expressions. The UPLC-MS outcome combining with network pharmacology identified mainly 20 active compounds in BJTD, with astragaloside IV exhibiting the strongest binding to the mTOR protein. The mTOR pathway modulation experiments confirmed that BJTD's protective effects are mediated through inhibition of hyperactivated mTOR phosphorylation and mitoch","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-03-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143555685","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}
Pub Date : 2025-02-22DOI: 10.1007/s10495-025-02083-3
Shansong Huang, Peicheng Ji, Peng Xu, Kanghui Liu, Han Ge, Zhengyuan Yan, Quan Cheng, Jialun Lv, Diancai Zhang
Peritoneal metastasis (PM) is the primary site of distant metastasis in gastric cancer (GC) and is associated with an advanced disease stage and poor prognosis. Due to its high resistance to chemotherapy, disseminated peritoneal lesions are often untreatable. A primary reason for therapy resistance in cancer cells is often their defective cell death execution mechanisms. Ferroptosis, a newly identified type of regulated cell death, is strongly linked to the emergence and formation of tumors. Earlier studies have demonstrated the significant role of RNA-binding proteins in ferroptosis. Nevertheless, the fundamental process linking Staufen Double-Stranded RNA Binding Protein 1 (STAU1) to ferroptosis in the peritoneal metastasis of gastric cancer is yet to be clarified. This study shows that the RNA-binding protein STAU1 is crucial for regulating ferroptosis in gastric cancer cells. Elevated levels of STAU1 are linked to unfavorable outcomes in individuals diagnosed with gastric cancer. STAU1 was up-regulated by PLAGL2 and decreased the stability of NCOA4 mRNA by binding to the 3ʹ-untranslated region. Decreased NCOA4 expression inhibits the accumulation of reactive iron, the occurrence of the Fenton reaction, and cellular ROS generation in the GC cells. Additionally, we showed that NCOA4 is crucial in the process of ferritinophagy triggered by the reduction of STAU1 in gastric cancer cells. Ultimately, the process safeguards GC cells from ferroptosis. These findings elucidate the function of PLAGL2/STAU1/NCOA4 in the ferroptosis of gastric cancer cells and provide theoretical backing for possible diagnostic markers and treatment targets for peritoneal metastasis in gastric cancer.
{"title":"PLAGL2-STAU1-NCOA4 axis enhances gastric cancer peritoneal metastasis by resisting ferroptosis via ferritinophagy","authors":"Shansong Huang, Peicheng Ji, Peng Xu, Kanghui Liu, Han Ge, Zhengyuan Yan, Quan Cheng, Jialun Lv, Diancai Zhang","doi":"10.1007/s10495-025-02083-3","DOIUrl":"10.1007/s10495-025-02083-3","url":null,"abstract":"<div><p>Peritoneal metastasis (PM) is the primary site of distant metastasis in gastric cancer (GC) and is associated with an advanced disease stage and poor prognosis. Due to its high resistance to chemotherapy, disseminated peritoneal lesions are often untreatable. A primary reason for therapy resistance in cancer cells is often their defective cell death execution mechanisms. Ferroptosis, a newly identified type of regulated cell death, is strongly linked to the emergence and formation of tumors. Earlier studies have demonstrated the significant role of RNA-binding proteins in ferroptosis. Nevertheless, the fundamental process linking Staufen Double-Stranded RNA Binding Protein 1 (STAU1) to ferroptosis in the peritoneal metastasis of gastric cancer is yet to be clarified. This study shows that the RNA-binding protein STAU1 is crucial for regulating ferroptosis in gastric cancer cells. Elevated levels of STAU1 are linked to unfavorable outcomes in individuals diagnosed with gastric cancer. STAU1 was up-regulated by PLAGL2 and decreased the stability of NCOA4 mRNA by binding to the 3ʹ-untranslated region. Decreased NCOA4 expression inhibits the accumulation of reactive iron, the occurrence of the Fenton reaction, and cellular ROS generation in the GC cells. Additionally, we showed that NCOA4 is crucial in the process of ferritinophagy triggered by the reduction of STAU1 in gastric cancer cells. Ultimately, the process safeguards GC cells from ferroptosis. These findings elucidate the function of PLAGL2/STAU1/NCOA4 in the ferroptosis of gastric cancer cells and provide theoretical backing for possible diagnostic markers and treatment targets for peritoneal metastasis in gastric cancer.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"30 3-4","pages":"1058 - 1075"},"PeriodicalIF":6.1,"publicationDate":"2025-02-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143476228","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}
The human body to prolonged high-altitude environments exposure can cause the decreased hepcidin and the disorder of iron homeostasis. Few animal studies have examined the molecular basis of iron metabolism disorder and the impact of hepcidin on the complex phenotype of high-altitude environments. In this study, we display reduced hepcidin level and impaired iron homeostasis upon hypobaric hypoxia, accompanied by mitochondrial dysfunction, abnormal blood cells and metabolic disorder. Importantly, mice overexpressing hepcidin show resistance to hypoxia-induced mitochondrial injury in liver and improve the body homeostasis. The comprehensive characterization of the metabolic pathways analysis demonstrates the porphyrin metabolism modulated by hepcidin may play a significant role in improving blood cell abnormalities upon high altitude hypoxia. In addition, hepcidin transcription is inhibited by histone modification and interruption of SMAD signaling pathway in hepatocytes in response to hypoxic stress to decrease hepcidin level. Our data highlight the key role of hepcidin in regulating the complex phenotype of high-altitude environments by altered metabolic and mitochondrial function. These results provide a theoretical reference to understanding the role of hepcidin in maintaining physiological function in high altitude, thus assisting in the development of strategies to better prevent and alleviate imbalance of energy homeostasis and adverse effects.
{"title":"Hepcidin mediates the disorder of iron homeostasis and mitochondrial function in mice under hypobaric hypoxia exposure","authors":"Jiayao Liu, Jialin Zhao, Jintao He, Yuhui Li, Jie Xu, Chenxi Xiao, Yuyu Zhang, Honghong Chen, Yajie Hu, Chunxiang Fan, Xinhua Liu","doi":"10.1007/s10495-025-02079-z","DOIUrl":"10.1007/s10495-025-02079-z","url":null,"abstract":"<div><p>The human body to prolonged high-altitude environments exposure can cause the decreased hepcidin and the disorder of iron homeostasis. Few animal studies have examined the molecular basis of iron metabolism disorder and the impact of hepcidin on the complex phenotype of high-altitude environments. In this study, we display reduced hepcidin level and impaired iron homeostasis upon hypobaric hypoxia, accompanied by mitochondrial dysfunction, abnormal blood cells and metabolic disorder. Importantly, mice overexpressing hepcidin show resistance to hypoxia-induced mitochondrial injury in liver and improve the body homeostasis. The comprehensive characterization of the metabolic pathways analysis demonstrates the porphyrin metabolism modulated by hepcidin may play a significant role in improving blood cell abnormalities upon high altitude hypoxia. In addition, hepcidin transcription is inhibited by histone modification and interruption of SMAD signaling pathway in hepatocytes in response to hypoxic stress to decrease hepcidin level. Our data highlight the key role of hepcidin in regulating the complex phenotype of high-altitude environments by altered metabolic and mitochondrial function. These results provide a theoretical reference to understanding the role of hepcidin in maintaining physiological function in high altitude, thus assisting in the development of strategies to better prevent and alleviate imbalance of energy homeostasis and adverse effects.</p></div>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":"30 3-4","pages":"1076 - 1091"},"PeriodicalIF":6.1,"publicationDate":"2025-02-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143476227","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}
Pub Date : 2025-02-20DOI: 10.1007/s10495-025-02086-0
Xianli Su, Yinhui Sun, Aiguo Dai
Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.
{"title":"New insights into pulmonary arterial hypertension: interaction between PANoptosis and perivascular inflammatory responses.","authors":"Xianli Su, Yinhui Sun, Aiguo Dai","doi":"10.1007/s10495-025-02086-0","DOIUrl":"https://doi.org/10.1007/s10495-025-02086-0","url":null,"abstract":"<p><p>Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.</p>","PeriodicalId":8062,"journal":{"name":"Apoptosis","volume":" ","pages":""},"PeriodicalIF":6.1,"publicationDate":"2025-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143466728","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}