Pub Date : 2026-01-09DOI: 10.1038/s41420-025-02859-1
Weijie Lu, Yaoting Deng, Mengyang Liu, Yujie Hu, Kang Yang, Bowen Wang, Yanling Li, Gang Wang, Li Ma, Jiajia Liu, Yaohui Xie, Qianrong Li, Ai Liu, Xiubin Zhang, Ping Xie
Acetylation is an important post-translational modification (PTM) of proteins and plays critical roles in multiple biological processes. The modes of cell death represent different pathways leading to the final outcome of "death" for the cell, with apoptosis, ferroptosis, and pyroptosis being the most common forms of cell death. In recent years, research has found that acetylation modifications influence various biological processes, ultimately playing a role in the regulation of apoptosis, ferroptosis, and pyroptosis. This article introduces the molecular effects of acetylation and enzyme/non-enzyme regulation, systematically summarizing the regulatory mechanisms of apoptosis, ferroptosis, and pyroptosis. It ultimately focuses on the genes and proteins associated with the regulation of apoptosis, ferroptosis, and pyroptosis, providing a comprehensive explanation of how acetylation regulates these processes. Given that the mode of cell death may be singular or coexist with two or more types in certain diseases, this article aims to conduct an in-depth analysis of the regulatory role of a specific PTM of proteins (acetylation) on different cell death pathways. Furthermore, it seeks to summarize potential key pathways or targets through which acetylation influences the interplay of various cell death mechanisms. By intervening in multiple cell death pathways, this study aims to provide insights for the prevention and treatment of tumors and cardiovascular diseases, both of which are closely related to outcomes associated with cell death.
{"title":"Regulation of apoptosis, ferroptosis, and pyroptosis mediated by acetylation.","authors":"Weijie Lu, Yaoting Deng, Mengyang Liu, Yujie Hu, Kang Yang, Bowen Wang, Yanling Li, Gang Wang, Li Ma, Jiajia Liu, Yaohui Xie, Qianrong Li, Ai Liu, Xiubin Zhang, Ping Xie","doi":"10.1038/s41420-025-02859-1","DOIUrl":"10.1038/s41420-025-02859-1","url":null,"abstract":"<p><p>Acetylation is an important post-translational modification (PTM) of proteins and plays critical roles in multiple biological processes. The modes of cell death represent different pathways leading to the final outcome of \"death\" for the cell, with apoptosis, ferroptosis, and pyroptosis being the most common forms of cell death. In recent years, research has found that acetylation modifications influence various biological processes, ultimately playing a role in the regulation of apoptosis, ferroptosis, and pyroptosis. This article introduces the molecular effects of acetylation and enzyme/non-enzyme regulation, systematically summarizing the regulatory mechanisms of apoptosis, ferroptosis, and pyroptosis. It ultimately focuses on the genes and proteins associated with the regulation of apoptosis, ferroptosis, and pyroptosis, providing a comprehensive explanation of how acetylation regulates these processes. Given that the mode of cell death may be singular or coexist with two or more types in certain diseases, this article aims to conduct an in-depth analysis of the regulatory role of a specific PTM of proteins (acetylation) on different cell death pathways. Furthermore, it seeks to summarize potential key pathways or targets through which acetylation influences the interplay of various cell death mechanisms. By intervening in multiple cell death pathways, this study aims to provide insights for the prevention and treatment of tumors and cardiovascular diseases, both of which are closely related to outcomes associated with cell death.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"15"},"PeriodicalIF":7.0,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12789129/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942656","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}
Pub Date : 2026-01-09DOI: 10.1038/s41420-025-02918-7
L Gaiaschi, F De Luca, C R Girelli, G Milanesi, E Roda, F P Fanizzi, M Grimaldi, M G Bottone
Glioblastoma stands as the deadliest primary brain malignancy in adults, primarily due to its resistance to conventional treatments and the restrictive nature of the blood-brain barrier (BBB). Cisplatin (CDDP), a widely used chemotherapeutic, demonstrates limited efficacy against glioblastoma owing to systemic toxicity and insufficient BBB penetration. To overcome these hurdles, we tested the platinum(II) complex [Pt(O,O'-acac)(γ-acac)(DMS)], indicated as Pt(acac)₂(DMS), known for its improved lipophilicity, ability to disrupt mitochondrial function, and reduced neurotoxic profile. Compared to CDDP, Pt(acac)₂(DMS) induced a targeted and prolonged cytotoxic response in U251 glioblastoma cells, promoting mitochondrial dysfunction, cell cycle arrest, and modulation of autophagy, while sparing primary human astrocytes. Our findings indicate that Pt(acac)₂(DMS) may overcome key limitations of cisplatin, including toxicity issues and resistance associated with autophagic adaptation, highlighting its promise as a potential therapeutic candidate for glioblastoma treatment.
{"title":"In vitro cytotoxic mechanisms of Pt(O,O'-acac)(γ-acac)(DMS): mitochondrial dysfunction and impaired autophagy in U251 cell line.","authors":"L Gaiaschi, F De Luca, C R Girelli, G Milanesi, E Roda, F P Fanizzi, M Grimaldi, M G Bottone","doi":"10.1038/s41420-025-02918-7","DOIUrl":"https://doi.org/10.1038/s41420-025-02918-7","url":null,"abstract":"<p><p>Glioblastoma stands as the deadliest primary brain malignancy in adults, primarily due to its resistance to conventional treatments and the restrictive nature of the blood-brain barrier (BBB). Cisplatin (CDDP), a widely used chemotherapeutic, demonstrates limited efficacy against glioblastoma owing to systemic toxicity and insufficient BBB penetration. To overcome these hurdles, we tested the platinum(II) complex [Pt(O,O'-acac)(γ-acac)(DMS)], indicated as Pt(acac)₂(DMS), known for its improved lipophilicity, ability to disrupt mitochondrial function, and reduced neurotoxic profile. Compared to CDDP, Pt(acac)₂(DMS) induced a targeted and prolonged cytotoxic response in U251 glioblastoma cells, promoting mitochondrial dysfunction, cell cycle arrest, and modulation of autophagy, while sparing primary human astrocytes. Our findings indicate that Pt(acac)₂(DMS) may overcome key limitations of cisplatin, including toxicity issues and resistance associated with autophagic adaptation, highlighting its promise as a potential therapeutic candidate for glioblastoma treatment.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":" ","pages":""},"PeriodicalIF":7.0,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942680","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}
Lung adenocarcinoma (LUAD) remains a major global health issue characterized by high incidence and mortality rates. RecQ-like helicase 4 (RECQL4), a member of the DNA helicase family, plays a crucial role in DNA replication, DNA damage repair, and tumor progression. However, its involvement and specific molecular mechanisms in LUAD progression have not been elucidated. Through this investigation, we found that RECQL4 expression was aberrantly elevated in clinical LUAD tissues, and higher levels of RECQL4 expression were associated with poor prognosis and worse clinicopathological characteristics in LUAD patients. Gain-of-function and loss-of-function studies demonstrated that RECQL4 promoted the proliferation, migration, and invasion abilities of LUAD cells. Subsequent gene set enrichment analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis confirmed that RECQL4 activates the NF-κB signaling pathway. Mechanistic investigation indicated that RECQL4 might function as a scaffold protein for the Y box binding protein 1 (YBX1) and GTPase-activating protein SH3 domain-binding protein 1 (G3BP1), enhancing the interaction between YBX1 and G3BP1, thereby activating the NF-κB signaling pathway and promoting the progression of LUAD. In conclusion, RECQL4 promotes the malignant progression of LUAD through the YBX1/G3BP1-mediated NF-κB signaling pathway. These findings suggest that RECQL4 has the potential to serve as a novel prognostic biomarker and an effective therapeutic target for LUAD.
{"title":"RECQL4 promotes the malignant progression of lung adenocarcinoma through the YBX1/G3BP1-mediated NF-κB signaling pathway.","authors":"Rongyang Li, Wenhao Yu, Dingxin Wang, Luyuan Ma, Zhanpeng Tang, Dingqiang Zhu, Zitong Feng, Wenqiang Qi, Hui Tian, Cun Gao","doi":"10.1038/s41420-025-02849-3","DOIUrl":"10.1038/s41420-025-02849-3","url":null,"abstract":"<p><p>Lung adenocarcinoma (LUAD) remains a major global health issue characterized by high incidence and mortality rates. RecQ-like helicase 4 (RECQL4), a member of the DNA helicase family, plays a crucial role in DNA replication, DNA damage repair, and tumor progression. However, its involvement and specific molecular mechanisms in LUAD progression have not been elucidated. Through this investigation, we found that RECQL4 expression was aberrantly elevated in clinical LUAD tissues, and higher levels of RECQL4 expression were associated with poor prognosis and worse clinicopathological characteristics in LUAD patients. Gain-of-function and loss-of-function studies demonstrated that RECQL4 promoted the proliferation, migration, and invasion abilities of LUAD cells. Subsequent gene set enrichment analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis confirmed that RECQL4 activates the NF-κB signaling pathway. Mechanistic investigation indicated that RECQL4 might function as a scaffold protein for the Y box binding protein 1 (YBX1) and GTPase-activating protein SH3 domain-binding protein 1 (G3BP1), enhancing the interaction between YBX1 and G3BP1, thereby activating the NF-κB signaling pathway and promoting the progression of LUAD. In conclusion, RECQL4 promotes the malignant progression of LUAD through the YBX1/G3BP1-mediated NF-κB signaling pathway. These findings suggest that RECQL4 has the potential to serve as a novel prognostic biomarker and an effective therapeutic target for LUAD.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"8"},"PeriodicalIF":7.0,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12789086/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942682","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}
Breast cancer (BC) is the most prevalent malignant disease affecting female patients globally, with triple-negative breast cancer (TNBC) being the subtype linked to the poorest clinical outcome. The liver is a frequent metastatic site of breast cancer. Therefore, elucidating the mechanism underlying liver metastasis in TNBC is crucial for identifying effective diagnostic and therapeutic targets, which holds significant potential for guiding clinical treatment. This study aimed to identify key genes driving breast cancer liver metastasis and to explore their functional mechanisms. Using RNA sequencing of metastatic 4T1-HM3 and primary 4T1-Pri tumor cells, mesothelin (MSLN) was identified as significantly upregulated in metastatic TNBC cells and tissues, as confirmed by qRT-PCR, Western blot, and immunohistochemistry. Further investigations revealed that MSLN overexpression is strongly correlated with liver metastasis compared to metastases at other sites. Mechanistically, MSLN binds to epidermal growth factor receptor (EGFR) and activates the EGFR-ERK1/2 signaling axis, thereby promoting TNBC cell survival and proliferation during metastasis. Importantly, targeting MSLN with a paclitaxel/carboplatin combination effectively inhibited liver metastasis of hepatotropic TNBC in a mouse model. Therefore, our study elucidates the role of the MSLN-mediated EGFR-ERK1/2 signaling pathway in TNBC liver metastasis and highlights potential targeted therapies for treating TNBC liver metastasis.
{"title":"MSLN-mediated activation of EGFR-ERK1/2 signaling drives liver metastasis in breast cancer.","authors":"Jing Chen, Zexiu Lu, Guowu Zhang, Die Meng, Chao Chang, Jian Chen, Boxuan Wang, Yanran Tong, Yuhang Hai, Ming Lei, Xingyu Yang, Yubi Gan, Chaoqun Deng, Peijin Dai, Manran Liu, Xi Tang","doi":"10.1038/s41420-025-02835-9","DOIUrl":"10.1038/s41420-025-02835-9","url":null,"abstract":"<p><p>Breast cancer (BC) is the most prevalent malignant disease affecting female patients globally, with triple-negative breast cancer (TNBC) being the subtype linked to the poorest clinical outcome. The liver is a frequent metastatic site of breast cancer. Therefore, elucidating the mechanism underlying liver metastasis in TNBC is crucial for identifying effective diagnostic and therapeutic targets, which holds significant potential for guiding clinical treatment. This study aimed to identify key genes driving breast cancer liver metastasis and to explore their functional mechanisms. Using RNA sequencing of metastatic 4T1-HM3 and primary 4T1-Pri tumor cells, mesothelin (MSLN) was identified as significantly upregulated in metastatic TNBC cells and tissues, as confirmed by qRT-PCR, Western blot, and immunohistochemistry. Further investigations revealed that MSLN overexpression is strongly correlated with liver metastasis compared to metastases at other sites. Mechanistically, MSLN binds to epidermal growth factor receptor (EGFR) and activates the EGFR-ERK1/2 signaling axis, thereby promoting TNBC cell survival and proliferation during metastasis. Importantly, targeting MSLN with a paclitaxel/carboplatin combination effectively inhibited liver metastasis of hepatotropic TNBC in a mouse model. Therefore, our study elucidates the role of the MSLN-mediated EGFR-ERK1/2 signaling pathway in TNBC liver metastasis and highlights potential targeted therapies for treating TNBC liver metastasis.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"11"},"PeriodicalIF":7.0,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12789440/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942703","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}
Pub Date : 2026-01-09DOI: 10.1038/s41420-025-02860-8
Xiaominting Song, Wenya Yang, Hang You, Shan Qian, Xiaoxue Hu, Ao Zhang, Jia Li, Yuzhi Li, Huachao Bin, Cheng Peng, Jin Pei, Zhixing Cao
The aryl hydrocarbon receptor (AHR) is a transcription factor prominently expressed at barrier sites, while aldehyde dehydrogenase 3 family member A1 (ALDH3A1) is a metabolic enzyme implicated in oxidative stress. However, their roles in ferroptosis remain poorly understood. Imperatorin (IMP) is a bioactive compound derived from traditional Chinese medicine. Here, we demonstrate that IMP is a natural agonist of AHR, inhibiting LPS-induced ferroptosis, inflammation, and barrier damage in lung epithelial cells by promoting AHR nuclear translocation and activation. Mechanistically, IMP-activated AHR stimulated the Nrf2/HO-1/GPX4 axis and enhanced ALDH3A1 expression, thereby inhibiting ferroptosis-related Fe2+ accumulation, ROS production, and lipid peroxidation. The in vivo results showed that oral IMP activated the AHR/ALDH3A1 and Nrf2/HO-1/GPX4 pathways in lung tissue, thus improving lung dysfunction and inflammation in acute lung injury (ALI) mice induced by LPS. Notably, ALDH3A1 is a key downstream signaling protein of AHR. An AHR inhibitor reversed the IMP-induced upregulation of ALDH3A1, whereas an ALDH3A1 inhibitor blocked the anti-ferroptotic Nrf2/HO-1/GPX4 pathway and diminished the lung-protective effects of IMP-activated AHR both in vitro and in vivo. These findings indicate that the AHR/ALDH3A1 axis may represent a previously unrecognized therapeutic target for ferroptosis and provide insight into IMP as a therapeutic strategy to prevent and treat ALI.
{"title":"ALDH3A1-dependent Nrf2/HO-1/GPX4 pathway supports AHR as a promising therapeutic target for ferroptosis and promotes imperatorin-mediated lung protection.","authors":"Xiaominting Song, Wenya Yang, Hang You, Shan Qian, Xiaoxue Hu, Ao Zhang, Jia Li, Yuzhi Li, Huachao Bin, Cheng Peng, Jin Pei, Zhixing Cao","doi":"10.1038/s41420-025-02860-8","DOIUrl":"10.1038/s41420-025-02860-8","url":null,"abstract":"<p><p>The aryl hydrocarbon receptor (AHR) is a transcription factor prominently expressed at barrier sites, while aldehyde dehydrogenase 3 family member A1 (ALDH3A1) is a metabolic enzyme implicated in oxidative stress. However, their roles in ferroptosis remain poorly understood. Imperatorin (IMP) is a bioactive compound derived from traditional Chinese medicine. Here, we demonstrate that IMP is a natural agonist of AHR, inhibiting LPS-induced ferroptosis, inflammation, and barrier damage in lung epithelial cells by promoting AHR nuclear translocation and activation. Mechanistically, IMP-activated AHR stimulated the Nrf2/HO-1/GPX4 axis and enhanced ALDH3A1 expression, thereby inhibiting ferroptosis-related Fe<sup>2+</sup> accumulation, ROS production, and lipid peroxidation. The in vivo results showed that oral IMP activated the AHR/ALDH3A1 and Nrf2/HO-1/GPX4 pathways in lung tissue, thus improving lung dysfunction and inflammation in acute lung injury (ALI) mice induced by LPS. Notably, ALDH3A1 is a key downstream signaling protein of AHR. An AHR inhibitor reversed the IMP-induced upregulation of ALDH3A1, whereas an ALDH3A1 inhibitor blocked the anti-ferroptotic Nrf2/HO-1/GPX4 pathway and diminished the lung-protective effects of IMP-activated AHR both in vitro and in vivo. These findings indicate that the AHR/ALDH3A1 axis may represent a previously unrecognized therapeutic target for ferroptosis and provide insight into IMP as a therapeutic strategy to prevent and treat ALI.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"16"},"PeriodicalIF":7.0,"publicationDate":"2026-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12789433/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145942738","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}
Recent studies have shown that heat shock protein 90 alpha family class A member 1 (HSP90AA1) interacts with various tumor-associated proteins, regulates their biological activity and stability, and plays an important role in various tumors. However, the role of HSP90AA1 in clear cell renal cell carcinoma (ccRCC) remains unclear. In the study, GEO and TCGA-KIRC databases were used to analyze the expression pattern and clinical significance of HSP90AA1 in ccRCC; immunohistochemistry and Western blot were used to validate HSP90AA1 expression in ccRCC tissues and cell lines; colony formation assays, EdU and TUNEL methods, cell migration and invasion experiments, and a mouse renal orthotopic xenograft tumor model were used to detect the effects of HSP90AA1 overexpression on the biological function of ccRCC; Co-IP and RNA-seq experiments were utilized to explore the downstream regulatory mechanism of HSP90AA1. Our results showed that HSP90AA1 expression was significantly downregulated in ccRCC, and its reduced expression was associated with tumor metastasis. HSP90AA1 overexpression markedly inhibited the proliferation and metastasis ability of ccRCC cells. HSP90AA1 bound to F-box only protein 7 (FBXO7) and accelerated its protein expression. FBXO7 was expressed at low level in ccRCC, and its decreased expression was closely related to unfavorable pathological features of tumors and poor patient prognosis. FBXO7 overexpression promoted cell adhesion molecule 1 (CADM1) expression and suppressed the PI3K-AKT signaling pathway. Knocking down FBXO7 expression on the basis of HSP90AA1 overexpression significantly reversed the cell phenotype inhibition caused by HSP90AA1 overexpression, downregulated CADM1 expression, and activated the PI3K-AKT signaling pathway. In summary, HSP90AA1 exhibited a low expression pattern in ccRCC, and HSP90AA1 overexpression promoted CADM1 expression and inhibited the PI3K-AKT pathway, thereby suppressing the proliferation and metastasis of ccRCC.
{"title":"HSP90AA1 restrains clear cell renal cell carcinoma progression by promoting CADM1 expression and suppressing the PI3K-AKT pathway through interaction with FBXO7.","authors":"Wuping Yang, Yifan Li, Zhi Li, Chaochao Jiang, Xu Deng, Ding Peng","doi":"10.1038/s41420-025-02848-4","DOIUrl":"10.1038/s41420-025-02848-4","url":null,"abstract":"<p><p>Recent studies have shown that heat shock protein 90 alpha family class A member 1 (HSP90AA1) interacts with various tumor-associated proteins, regulates their biological activity and stability, and plays an important role in various tumors. However, the role of HSP90AA1 in clear cell renal cell carcinoma (ccRCC) remains unclear. In the study, GEO and TCGA-KIRC databases were used to analyze the expression pattern and clinical significance of HSP90AA1 in ccRCC; immunohistochemistry and Western blot were used to validate HSP90AA1 expression in ccRCC tissues and cell lines; colony formation assays, EdU and TUNEL methods, cell migration and invasion experiments, and a mouse renal orthotopic xenograft tumor model were used to detect the effects of HSP90AA1 overexpression on the biological function of ccRCC; Co-IP and RNA-seq experiments were utilized to explore the downstream regulatory mechanism of HSP90AA1. Our results showed that HSP90AA1 expression was significantly downregulated in ccRCC, and its reduced expression was associated with tumor metastasis. HSP90AA1 overexpression markedly inhibited the proliferation and metastasis ability of ccRCC cells. HSP90AA1 bound to F-box only protein 7 (FBXO7) and accelerated its protein expression. FBXO7 was expressed at low level in ccRCC, and its decreased expression was closely related to unfavorable pathological features of tumors and poor patient prognosis. FBXO7 overexpression promoted cell adhesion molecule 1 (CADM1) expression and suppressed the PI3K-AKT signaling pathway. Knocking down FBXO7 expression on the basis of HSP90AA1 overexpression significantly reversed the cell phenotype inhibition caused by HSP90AA1 overexpression, downregulated CADM1 expression, and activated the PI3K-AKT signaling pathway. In summary, HSP90AA1 exhibited a low expression pattern in ccRCC, and HSP90AA1 overexpression promoted CADM1 expression and inhibited the PI3K-AKT pathway, thereby suppressing the proliferation and metastasis of ccRCC.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"6"},"PeriodicalIF":7.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783301/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932332","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}
Pub Date : 2026-01-08DOI: 10.1038/s41420-025-02930-x
Wenjun Wang, Yibiao Shi, Sitian Qiu, Ying Song, Xi Chen, Xiaomin Zhang, Beibei Wang, Qisong Li, Qiwen Shi
In the present study, we aimed to investigate the antioxidant and therapeutic protective effects of carvacryl acetate (CAA) on Mitochondrial damage of cerebral ischemia-reperfusion through mitochondrial transcription factor A (TFAM) signaling molecules.SD rats were used to establish the middle cerebral artery occlusion (MCAO) model in vivo, and PC12 cells were stimulated with H2O2 in vitro. Longa neurological score and triphenyltetrazolium chloride (TTC) staining was used to observe the ischemic infarction. Transmission electron microscope (TEM) was used to observe the mitochondria. Reactive Oxygen Species/ Superoxide Dismuptase/Malondialdehyde/Adenosine Triphosphate (ROS/SOD/MDA/ATP) detection kit was used to detect. RT-qPCR was used to detect the mRNA level of target gene and mitochondrial DNA (mtDNA) copy number changes. Immunofluorescence and Western blot were used to detect the expression of protein. After oxidative stress in the MCAO model of SD rats, the neurological score increased, the volume of ischemic area of cerebral infarction increased, the morphology of nerve cells in brain tissue and PC12 cells was disordered, the mitochondria appeared vacuolated, the contents of ROS and MDA increased, and the activity of SOD decreased. Oxidative stress causes mitochondrial dysfunction, resulting in the reduction of mtDNA copy number and the decreased expression of TFAM in brain tissue nerve cells and PC12 cells, which in turn affects mitochondrial transcription biogenesis and decreases the expression of POLRMT and TFB2M molecules. CAA promotes intracellular TFAM expression and activates its antioxidant pathway, thereby protecting mtDNA and alleviating oxidative stress and mitochondrial damage caused by MCAO in vivo and H2O2 stimulation in vitro. Lentivirus downregulates the expression of TFAM, and under its action, the antioxidant and mitochondrial protection effects of CAA are weakened. When TFAM was disrupted, the protective effect of CAA on mitochondria was inhibited. Compared to edaravone, a positive control, CAA exhibited similar therapeutic effects. These findings suggest that CAA alleviates CIRI through TFAM signaling pathways, offering potential therapeutic implications for ischemic stroke treatment.
{"title":"TFAM signaling molecule alleviates mitochondrial damage of cerebral ischemia-reperfusion.","authors":"Wenjun Wang, Yibiao Shi, Sitian Qiu, Ying Song, Xi Chen, Xiaomin Zhang, Beibei Wang, Qisong Li, Qiwen Shi","doi":"10.1038/s41420-025-02930-x","DOIUrl":"https://doi.org/10.1038/s41420-025-02930-x","url":null,"abstract":"<p><p>In the present study, we aimed to investigate the antioxidant and therapeutic protective effects of carvacryl acetate (CAA) on Mitochondrial damage of cerebral ischemia-reperfusion through mitochondrial transcription factor A (TFAM) signaling molecules.SD rats were used to establish the middle cerebral artery occlusion (MCAO) model in vivo, and PC12 cells were stimulated with H<sub>2</sub>O<sub>2</sub> in vitro. Longa neurological score and triphenyltetrazolium chloride (TTC) staining was used to observe the ischemic infarction. Transmission electron microscope (TEM) was used to observe the mitochondria. Reactive Oxygen Species/ Superoxide Dismuptase/Malondialdehyde/Adenosine Triphosphate (ROS/SOD/MDA/ATP) detection kit was used to detect. RT-qPCR was used to detect the mRNA level of target gene and mitochondrial DNA (mtDNA) copy number changes. Immunofluorescence and Western blot were used to detect the expression of protein. After oxidative stress in the MCAO model of SD rats, the neurological score increased, the volume of ischemic area of cerebral infarction increased, the morphology of nerve cells in brain tissue and PC12 cells was disordered, the mitochondria appeared vacuolated, the contents of ROS and MDA increased, and the activity of SOD decreased. Oxidative stress causes mitochondrial dysfunction, resulting in the reduction of mtDNA copy number and the decreased expression of TFAM in brain tissue nerve cells and PC12 cells, which in turn affects mitochondrial transcription biogenesis and decreases the expression of POLRMT and TFB<sub>2</sub>M molecules. CAA promotes intracellular TFAM expression and activates its antioxidant pathway, thereby protecting mtDNA and alleviating oxidative stress and mitochondrial damage caused by MCAO in vivo and H<sub>2</sub>O<sub>2</sub> stimulation in vitro. Lentivirus downregulates the expression of TFAM, and under its action, the antioxidant and mitochondrial protection effects of CAA are weakened. When TFAM was disrupted, the protective effect of CAA on mitochondria was inhibited. Compared to edaravone, a positive control, CAA exhibited similar therapeutic effects. These findings suggest that CAA alleviates CIRI through TFAM signaling pathways, offering potential therapeutic implications for ischemic stroke treatment.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":" ","pages":""},"PeriodicalIF":7.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932353","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}
Prostate cancer (PCa) patients with bone metastasis commonly exhibit osteoblastic-type and have an extremely poor prognosis. Exosomes derived from tumor cells possess biological significance and can mediate intercellular communication in the tumor microenvironment. Long noncoding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) is implicated in tumorigenesis and the development of PCa, but the precise roles of SNHG1 in the regulation of bone homeostasis remain elusive. Herein, we aimed to investigate the underlying mechanisms by which exosomes-encapsulated SNHG1 affects the bone metastasis of PCa. Our findings revealed that SNHG1 was overexpressed in PCa tissues, highly enriched in PCa cell-derived exosomes, and positively correlated with bone metastasis. Besides, SNHG1 shuttled by PCa-derived exosomes could be transferred into osteoblast cells, where SNHG1 exerted inductive properties in osteogenic differentiation. Gain- and loss-of-functional experiments demonstrated that exosomal SNHG1 facilitated the activity of alkaline phosphatase and mineralization of extracellular matrix. Moreover, in vivo experimentation showed that knockdown of exosomal SNHG1 suppressed bone metastasis of PCa cells. Mechanistic investigations revealed that exosomal SNHG1, transmitted to osteoblast cells, physically binds to YBX1 and leads to the shift of YBX1 into the nucleus, then enhances MMP16 transcription and increases the amount of protein translation, ultimately resulting in PCa bone metastasis. In conclusion, our data highlight that PCa-derived exosomes-loaded SNHG1 mediated osteogenesis through the SNHG1/YBX1/MMP16 axis. SNHG1 may serve as a potential diagnostic marker and therapeutic target for bone metastasis in PCa.
{"title":"Exosome-transmitted long noncoding RNA SNHG1 promotes prostate cancer bone metastasis via YBX1/MMP16 axis.","authors":"Taowei Yang, Junqi Luo, Zining Long, Jun Wu, Wenbin Chen, Xumin Zhou, Libin Zou, Shengren Cen, Chuanfan Zhong, Jianming Lu, Pengxiang Zheng, Anyang Wei, Daojun Lv, Xiangming Mao","doi":"10.1038/s41420-025-02855-5","DOIUrl":"10.1038/s41420-025-02855-5","url":null,"abstract":"<p><p>Prostate cancer (PCa) patients with bone metastasis commonly exhibit osteoblastic-type and have an extremely poor prognosis. Exosomes derived from tumor cells possess biological significance and can mediate intercellular communication in the tumor microenvironment. Long noncoding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) is implicated in tumorigenesis and the development of PCa, but the precise roles of SNHG1 in the regulation of bone homeostasis remain elusive. Herein, we aimed to investigate the underlying mechanisms by which exosomes-encapsulated SNHG1 affects the bone metastasis of PCa. Our findings revealed that SNHG1 was overexpressed in PCa tissues, highly enriched in PCa cell-derived exosomes, and positively correlated with bone metastasis. Besides, SNHG1 shuttled by PCa-derived exosomes could be transferred into osteoblast cells, where SNHG1 exerted inductive properties in osteogenic differentiation. Gain- and loss-of-functional experiments demonstrated that exosomal SNHG1 facilitated the activity of alkaline phosphatase and mineralization of extracellular matrix. Moreover, in vivo experimentation showed that knockdown of exosomal SNHG1 suppressed bone metastasis of PCa cells. Mechanistic investigations revealed that exosomal SNHG1, transmitted to osteoblast cells, physically binds to YBX1 and leads to the shift of YBX1 into the nucleus, then enhances MMP16 transcription and increases the amount of protein translation, ultimately resulting in PCa bone metastasis. In conclusion, our data highlight that PCa-derived exosomes-loaded SNHG1 mediated osteogenesis through the SNHG1/YBX1/MMP16 axis. SNHG1 may serve as a potential diagnostic marker and therapeutic target for bone metastasis in PCa.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"7"},"PeriodicalIF":7.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783806/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932288","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}
Pub Date : 2026-01-08DOI: 10.1038/s41420-025-02822-0
Rong Zhang, Shengjun Chai, Fengjuan Zhang, Jiaming Lai, Rui Wang, Guocai Xu, Xiaoxia Fan, Botong Li, Chunmei Cai
This study examines hypoxia's role in regulating ATXN3 (ATXN3) across cervical cancer subtypes and its impact on tumor progression. We analyzed ATXN3 expression in clinical samples and cell lines (C33A, HeLa, SiHa), assessing proliferation/migration/invasion after ATXN3 modulation. The study investigated whether ATXN3 is regulated by hypoxia through hypoxia-inducible factor 1α (HIF-1α). Downstream mechanisms were explored using clinical samples and cell lines, comparing P53 and signal transducer and activator of transcription 5 (STAT5)/p-STAT5 levels between cancer tissues and adjacent non-cancerous tissues, and assessing changes following ATXN3 manipulation. ATXN3 was downregulated in human papillomavirus(HPV18+) cervical adenocarcinoma but upregulated in HPV16+ cervical squamous cell carcinoma. ATXN3 suppressed malignant behaviors in C33A and HeLa but promoted them in SiHa. HIF-1α expression was elevated in cancer tissues versus non-cancerous tissues, with hypoxic conditions differentially regulating ATXN3 via HIF-1α across cell lines. Cervical cancer tissues showed lower P53 and higher p-STAT5 (in HPV16+ squamous cell carcinoma). ATXN3 overexpression stabilized P53 in C33A/HeLa and increased p-STAT5 in SiHa, with inverse effects upon silencing. The findings suggest that hypoxia promotes the progression of subtypes of cervical cancer by regulating ATXN3-enhanced P53/p-STAT5 levels, which may provide a novel therapeutic strategy for clinical applications.
{"title":"Hypoxia promotes progression of cervical cancer by modulating the ATXN3-enhanced P53 stability or STAT5 phosphorylation.","authors":"Rong Zhang, Shengjun Chai, Fengjuan Zhang, Jiaming Lai, Rui Wang, Guocai Xu, Xiaoxia Fan, Botong Li, Chunmei Cai","doi":"10.1038/s41420-025-02822-0","DOIUrl":"10.1038/s41420-025-02822-0","url":null,"abstract":"<p><p>This study examines hypoxia's role in regulating ATXN3 (ATXN3) across cervical cancer subtypes and its impact on tumor progression. We analyzed ATXN3 expression in clinical samples and cell lines (C33A, HeLa, SiHa), assessing proliferation/migration/invasion after ATXN3 modulation. The study investigated whether ATXN3 is regulated by hypoxia through hypoxia-inducible factor 1α (HIF-1α). Downstream mechanisms were explored using clinical samples and cell lines, comparing P53 and signal transducer and activator of transcription 5 (STAT5)/p-STAT5 levels between cancer tissues and adjacent non-cancerous tissues, and assessing changes following ATXN3 manipulation. ATXN3 was downregulated in human papillomavirus(HPV18<sup>+</sup>) cervical adenocarcinoma but upregulated in HPV16<sup>+</sup> cervical squamous cell carcinoma. ATXN3 suppressed malignant behaviors in C33A and HeLa but promoted them in SiHa. HIF-1α expression was elevated in cancer tissues versus non-cancerous tissues, with hypoxic conditions differentially regulating ATXN3 via HIF-1α across cell lines. Cervical cancer tissues showed lower P53 and higher p-STAT5 (in HPV16+ squamous cell carcinoma). ATXN3 overexpression stabilized P53 in C33A/HeLa and increased p-STAT5 in SiHa, with inverse effects upon silencing. The findings suggest that hypoxia promotes the progression of subtypes of cervical cancer by regulating ATXN3-enhanced P53/p-STAT5 levels, which may provide a novel therapeutic strategy for clinical applications.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"4"},"PeriodicalIF":7.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783129/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932357","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}
Pub Date : 2026-01-08DOI: 10.1038/s41420-025-02851-9
Yuwen Chen, Ming Yang, Wenhui Xie, Huashan Hong
Hypertension is a highly prevalent chronic disease all around the world, and the pathogenic mechanism is complicated. The early and rapid decline of the function of human vascular system due to the aging of human body are characteristics of hypertension, which is accompanied by progressive pathological remodeling and arterial stiffening. The pathogenetic action of oxidation and inflammation is the vital function in the process of endothelial dysfunction and arterial injury. Bone marrow is considered as the birthplace of the immune cell, and the role of bone marrow in hematopoiesis and immune response for the onset of hypertension has been confirmed. In turn, inflammatory and oxidative stress also affect the bone marrow and damage bone marrow function, causing a series of complications in hypertension, resulting in a vicious cycle. Recently, increasing evidence has suggested that bone marrow aging plays an important role in the onset and development of hypertension, and that the function of bone marrow in the pathogenesis of hypertension has been seriously overlooked. Bone marrow microvascular ageing is also involved in the progression of bone marrow ageing. Thus, this review mainly focuses on bone marrow function in aging and hypertension progression, addresses the current studies on the roles of vascular aging, the bone marrow and the immune system in hypertension, and discusses their interaction and function in the pathogenesis of hypertension. Furthermore, some novel molecular pathological mechanisms are surveyed. This can add a new impetus to the mechanism research of hypertension onset.
{"title":"The role of vascular aging, bone marrow and immune system in hypertension.","authors":"Yuwen Chen, Ming Yang, Wenhui Xie, Huashan Hong","doi":"10.1038/s41420-025-02851-9","DOIUrl":"10.1038/s41420-025-02851-9","url":null,"abstract":"<p><p>Hypertension is a highly prevalent chronic disease all around the world, and the pathogenic mechanism is complicated. The early and rapid decline of the function of human vascular system due to the aging of human body are characteristics of hypertension, which is accompanied by progressive pathological remodeling and arterial stiffening. The pathogenetic action of oxidation and inflammation is the vital function in the process of endothelial dysfunction and arterial injury. Bone marrow is considered as the birthplace of the immune cell, and the role of bone marrow in hematopoiesis and immune response for the onset of hypertension has been confirmed. In turn, inflammatory and oxidative stress also affect the bone marrow and damage bone marrow function, causing a series of complications in hypertension, resulting in a vicious cycle. Recently, increasing evidence has suggested that bone marrow aging plays an important role in the onset and development of hypertension, and that the function of bone marrow in the pathogenesis of hypertension has been seriously overlooked. Bone marrow microvascular ageing is also involved in the progression of bone marrow ageing. Thus, this review mainly focuses on bone marrow function in aging and hypertension progression, addresses the current studies on the roles of vascular aging, the bone marrow and the immune system in hypertension, and discusses their interaction and function in the pathogenesis of hypertension. Furthermore, some novel molecular pathological mechanisms are surveyed. This can add a new impetus to the mechanism research of hypertension onset.</p>","PeriodicalId":9735,"journal":{"name":"Cell Death Discovery","volume":"12 1","pages":"3"},"PeriodicalIF":7.0,"publicationDate":"2026-01-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12783719/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145932267","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}