Cell Death and Differentiation最新文献

筛选
英文 中文
PDK4-driven lactate accumulation facilitates LPCAT2 lactylation to exacerbate sepsis-induced acute lung injury. pdk4驱动的乳酸积累促进LPCAT2的乳酸化,加剧败血症引起的急性肺损伤。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-10-07 DOI: 10.1038/s41418-025-01585-6
Yifan Deng, Yuetan Qiu, Xiang Li, Ting Gong, Jinyan Guo, Haoxuan Liang, Ziyi Yuan, Ziqing Hei, Xuedi Zhang, Youtan Liu
{"title":"PDK4-driven lactate accumulation facilitates LPCAT2 lactylation to exacerbate sepsis-induced acute lung injury.","authors":"Yifan Deng, Yuetan Qiu, Xiang Li, Ting Gong, Jinyan Guo, Haoxuan Liang, Ziyi Yuan, Ziqing Hei, Xuedi Zhang, Youtan Liu","doi":"10.1038/s41418-025-01585-6","DOIUrl":"https://doi.org/10.1038/s41418-025-01585-6","url":null,"abstract":"<p><p>Elevated glycolysis in lung tissue is a hallmark of sepsis-induced acute lung injury (SI-ALI), yet the role of glycolytic reprogramming and lactate-derived protein modifications in damaging epithelial cells remains poorly understood. In this study, we reveal that PDK4-driven glycolytic reprogramming promotes excessive lactate production in lung tissue during SI-ALI. Mechanistically, AARS1 in epithelial cells selectively enhances lactylation modification at the K375 site of LPCAT2, which suppresses STAT1 acetylation and facilitates STAT1 phosphorylation, nuclear translocation, and transcriptional repression of SLC7A11. This cascade ultimately triggers epithelial cells ferroptosis. Pharmacological inhibition of PDK4 attenuates lactate accumulation and LPCAT2 lactylation, thereby restoring STAT1 acetylation and SLC7A11 expression. Furthermore, AARS1 knockdown or mutation of the LPCAT2-K375 lactylation site rescues STAT1-mediated SLC7A11 suppression and mitigates ferroptosis in vitro and septic mice. Our findings revealed that elevated expression of PDK4 is a critical factor contributing to the increased lactate production in lung tissue during sepsis, and established a novel LPCAT2-K375/STAT1/SLC7A11 axis driving epithelial cells ferroptosis in SI-ALI, highlighting the crosstalk between metabolic reprogramming, post-translational modifications (PTM), and ferroptosis. Targeting the PDK4 or LPCAT2 lactylation may offer therapeutic potential for SI-ALI. In sepsis-induced acute lung injury (SI-ALI), PDK4 hyperactivation drives excessive lactate production in epithelial cells, triggering AARS1/HDAC9-mediated LPCAT2 lactylation. This modification suppresses STAT1 acetylation while enhancing phosphorylation, driving its nuclear translocation and subsequent SLC7A11 transcriptional downregulation. The resultant glutathione synthesis deficiency promotes ferroptosis, exacerbating SI-ALI progression.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145243855","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Caspase-8 expression and its Src dependent phosphorylation on Tyrosine 380 triggers NRF2 signaling activation in glioblastoma. 在胶质母细胞瘤中,Caspase-8的表达及其Src依赖的酪氨酸380磷酸化可触发NRF2信号激活。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-10-06 DOI: 10.1038/s41418-025-01542-3
Claudia Cirotti, Claudia Di Girolamo, Irene Taddei, Claudia Contadini, Giorgia Massacci, Francesca Sacco, Donatella Del Bufalo, Illari Salvatori, Cristiana Valle, Daniela Barilà
{"title":"Caspase-8 expression and its Src dependent phosphorylation on Tyrosine 380 triggers NRF2 signaling activation in glioblastoma.","authors":"Claudia Cirotti, Claudia Di Girolamo, Irene Taddei, Claudia Contadini, Giorgia Massacci, Francesca Sacco, Donatella Del Bufalo, Illari Salvatori, Cristiana Valle, Daniela Barilà","doi":"10.1038/s41418-025-01542-3","DOIUrl":"https://doi.org/10.1038/s41418-025-01542-3","url":null,"abstract":"<p><p>Caspase-8 expression is upregulated in many tumors where, despite its canonical apoptotic pathway, it sustains cancer progression promoting cell migration, NF-kB activation and inflammation. Here, we provide the first evidence for a novel role of Caspase-8 in promoting the metabolic rewiring of cancer cells. By performing transcriptomic, proteomic and phosphoproteomic analyses on glioblastoma cellular models, we identify Caspase-8 as an unexpected modulator of NRF2. Here we show that Caspase-8 expression and phosphorylation affect NRF2 activity and mitochondrial homeostasis. Mechanistically, we demonstrate that Src-dependent phosphorylation of Caspase-8 on Tyrosine 380 (Y380), frequently reported in cancers including glioblastoma, sustains mTORC1 activation, thus promoting energy metabolism. mTORC1 activity results in p62 phosphorylation allowing its dependent sequestration of KEAP1 protein and constitutive NRF2 signaling activation, as a consequence. Overall, this work depicted a novel unexpected role for Caspase-8 in the modulation of cancer cell metabolism, bridging together Src, mTORC1 and NRF2 signaling.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145238144","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
O-GlcNAcylation of UGDH regulates its activity and remodels the extracellular matrix to facilitate tumor growth. UGDH的o - glcn酰化调节其活性,重塑细胞外基质,促进肿瘤生长。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-10-06 DOI: 10.1038/s41418-025-01591-8
Bingyi Lin, Junjie Zhou, Didi Geng, Siyuan Chai, Xuanming Zhang, Zengle Zhang, Jiating Hu, Qin Tang, Xiaoming Chen, Wen Yi, Liming Wu
{"title":"O-GlcNAcylation of UGDH regulates its activity and remodels the extracellular matrix to facilitate tumor growth.","authors":"Bingyi Lin, Junjie Zhou, Didi Geng, Siyuan Chai, Xuanming Zhang, Zengle Zhang, Jiating Hu, Qin Tang, Xiaoming Chen, Wen Yi, Liming Wu","doi":"10.1038/s41418-025-01591-8","DOIUrl":"https://doi.org/10.1038/s41418-025-01591-8","url":null,"abstract":"<p><p>The tumor microenvironment is an immunosuppressive niche that contributes to tumor growth by downregulating immune cell functions or restraining immune cell infiltration. The underlying mechanisms are not still poorly understood. Here, we demonstrate that O-linked N-acetylglucosamine (O-GlcNAcylation), a prevalent form of protein glycosylation, contributes to establishing the immunosuppressive niche through regulating the metabolic and non-metabolic functions of uridine diphosphate glucose dehydrogenase (UGDH). Tumor cells carrying O-GlcNAcylation-deficient UGDH showed reduced xenograft tumor growth and improved survival in mice. Cytometry by time-of-flight (CyTOF) analysis suggests UGDH O-GlcNAcylation negatively correlates with cytotoxic CD8<sup>+</sup> T cell infiltration. O-GlcNAcylation on serine 350 of UGDH is located within the UDP-binding domain, and the subsequent extensive all-atom molecular dynamics simulations reveal that O-GlcNAcylation reinforces hydrogen-bonding interaction and enzymatic activity of UGDH, leading to enhanced hyaluronic acid (HA) synthesis in the extracellular matrix. Moreover, O-GlcNAcylation of UGDH reduces CD8<sup>+</sup> T cell infiltration by decreasing the chemokine CXCL10 expression. Specifically, O-GlcNAcylation enhances UGDH interaction with KPNA2 to compete with STAT1, and suppresses translocation of STAT1 into the nucleus, thereby transcriptionally downregulating CXCL10 expression. Thus, our study identifies UGDH O-GlcNAcylation as a key regulator of tumor immunity and further suggests a potential strategy for enhancing immunotherapy.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145238199","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
K11- and K29-ubiquitination-mediated nuclear translocation of glycolytic enzyme aldolase A promotes pancreatic cancer progression by NF-κB activation. K11-和k29泛素化介导的糖酵解酶醛缩酶A核易位通过NF-κ b活化促进胰腺癌进展。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-10-04 DOI: 10.1038/s41418-025-01592-7
Siru Zhou, Yulin Li, Chao Wang, Yuhan Zhao, Xiaofeng Zheng
{"title":"K11- and K29-ubiquitination-mediated nuclear translocation of glycolytic enzyme aldolase A promotes pancreatic cancer progression by NF-κB activation.","authors":"Siru Zhou, Yulin Li, Chao Wang, Yuhan Zhao, Xiaofeng Zheng","doi":"10.1038/s41418-025-01592-7","DOIUrl":"https://doi.org/10.1038/s41418-025-01592-7","url":null,"abstract":"<p><p>The function of cytosolic aldolase A (ALDOA) in glycolysis is well recognized. However, the cytosol-to-nucleus redistribution of ALDOA and its nuclear function is poorly understood. Here, we uncover inflammatory factor-stimulated nuclear function of ALDOA in augmenting pancreatic carcinogenesis by activating NF-κB signaling in a ubiquitination-dependent manner. TNF-α-triggered K11- and K29-linked ubiquitination of ALDOA at Lys200 promotes its interaction with RelA/p65 and facilitates importin-β-dependent nuclear translocation, establishing a positive feedback regulation in the tumor microenvironment by elevating the TNF-α expression in pancreatic ductal adenocarcinoma (PDAC). USP4 is identified as a negative regulator that deubiquitinates ALDOA. Instead of broadly targeting ALDOA, which causes glycolysis impairment, the specific elimination of ALDOA ubiquitination enhances chemosensitivity and the synergistic effect of chemotherapy combined with p65-specific anti-inflammatory therapy by selectively suppressing inflammation-induced proliferation in cancer cells. Collectively, we unveil the multifaceted mechanisms by which ALDOA promotes PDAC carcinogenesis, from metabolic to gene regulatory perspectives, providing potential therapies combatting cancer.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145228414","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neuronal MCT2 promotes angiogenesis via lactate in the developing mouse neocortex. 神经元MCT2通过乳酸促进发育中的小鼠新皮层的血管生成。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-10-04 DOI: 10.1038/s41418-025-01581-w
Daehoon Lee, Anika Wu, Lingling Yao, Shreya Satish, Lin Mei, Wen-Cheng Xiong
{"title":"Neuronal MCT2 promotes angiogenesis via lactate in the developing mouse neocortex.","authors":"Daehoon Lee, Anika Wu, Lingling Yao, Shreya Satish, Lin Mei, Wen-Cheng Xiong","doi":"10.1038/s41418-025-01581-w","DOIUrl":"https://doi.org/10.1038/s41418-025-01581-w","url":null,"abstract":"<p><p>Neural activity drives blood vessel (BV) formation and energy substrate delivery in the developing brain to meet rising metabolic demands; however, the underlying mechanisms remain poorly understood. In this study, we exposed neonatal mice to chronic whisker stimulation (WS), a paradigm known to enhance BV formation in the somatosensory (S1) cortex. Transcriptomic (RNA-seq) and spatial (RNA-scope) analyses revealed that WS upregulated monocarboxylate transporter 2 (MCT2) in cortical neurons and MCT1 in endothelial cells (ECs). These changes coincided with increased cortical lactate levels, elevated astrocytic vascular endothelial growth factor A (VEGFa), and enhanced angiogenesis. Functional experiments demonstrated that neuronal MCT2 is essential for mediating WS-induced angiogenic and metabolic responses. Mechanistically, MCT2 facilitates <sub>L</sub>-lactate influx into the cortex with or without WS, promoting lactate uptake by neurons and astrocytes. This, in turn, induces MCT2 expression in neurons and activates hypoxia-inducible factor 1α (HIF1α) and VEGFa expression in astrocytes. Together, these findings uncover a previously unrecognized role for neuronal MCT2 in regulating lactate flux, signaling, and vascular remodeling, thereby linking neural activity to metabolic adaptation and vascular development in the neonatal mouse neocortex.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-10-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145228381","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The legacy of a gentleman scientist: Pierre Hainaut. 一位绅士科学家的遗产:皮埃尔·海诺。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-10-03 DOI: 10.1038/s41418-025-01586-5
Christophe Arnoult, Laura D Attardi, Kerem Batsheva, Giovanni Blandino, Kathleen H Burns, Giannino Del Sal, David G Kirsch, David P Lane, Arnold J Levine, Guillermina Lozano, David Malkin, Gerry Melino, Moshe Oren, Carol Prives, Daniel Schramek
{"title":"The legacy of a gentleman scientist: Pierre Hainaut.","authors":"Christophe Arnoult, Laura D Attardi, Kerem Batsheva, Giovanni Blandino, Kathleen H Burns, Giannino Del Sal, David G Kirsch, David P Lane, Arnold J Levine, Guillermina Lozano, David Malkin, Gerry Melino, Moshe Oren, Carol Prives, Daniel Schramek","doi":"10.1038/s41418-025-01586-5","DOIUrl":"https://doi.org/10.1038/s41418-025-01586-5","url":null,"abstract":"","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145225183","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
AMPK-activated BAP1 regulates pVHL stability and tumor-suppressive functions. ampk激活的BAP1调节pVHL的稳定性和肿瘤抑制功能。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-09-27 DOI: 10.1038/s41418-025-01590-9
Mei Li, Lei Huang, Jiayi Chen, Tangming Guan, Yalei Wen, Yingjie Zhu, Xiao Yang, Caishi Zhang, Xiuqing Ma, Rui Wan, Yuanqiao He, Yang Zhou, Yan Song, Haoxing Zhang, Tongzheng Liu
{"title":"AMPK-activated BAP1 regulates pVHL stability and tumor-suppressive functions.","authors":"Mei Li, Lei Huang, Jiayi Chen, Tangming Guan, Yalei Wen, Yingjie Zhu, Xiao Yang, Caishi Zhang, Xiuqing Ma, Rui Wan, Yuanqiao He, Yang Zhou, Yan Song, Haoxing Zhang, Tongzheng Liu","doi":"10.1038/s41418-025-01590-9","DOIUrl":"https://doi.org/10.1038/s41418-025-01590-9","url":null,"abstract":"<p><p>The von Hippel-Lindau (VHL) protein (pVHL) functions as a potent tumor suppressor by mediating the degradation or inactivation of various substrates, including HIFα and Akt. However, pVHL is frequently downregulated in numerous cancers harboring wild-type VHL, and underlying mechanisms remains elusive. Aberrant glucose metabolism is a hallmark of cancer, driving tumor progression and therapeutic resistance. Despite this, the connection between glucose homoeostasis and pVHL turnover and functions has yet to be defined. In this study, we demonstrate that dysregulated glucose metabolism destabilizes pVHL in pancreatic ductal adenocarcinoma (PDAC), colorectal, and ovarian cancer cells. Mechanistically, energy stress induced by glucose starvation, 2-deoxyglucose (2-DG), or metformin activates AMP-activated protein kinase (AMPK), which subsequently phosphorylates and activates BAP1, a deubiquitinase whose specific function in targeting pVHL for deubiquitination and stabilization had not been previously characterized. Specifically, AMPKα phosphorylates BAP1 at residues S123, S469, and S583, enhancing the interaction between BAP1 and pVHL and promoting pVHL stabilization and tumor-suppressive function both in vitro and in vivo. Conversely, disrupting BAP1 phosphorylation through AMPKα depletion or reconstitution with a phosphorylation-defective BAP1 mutant (S123A/S469A/S583A) abolishes the BAP1-pVHL interaction, leading to impaired pVHL stabilization and accelerated tumor progression in cancer cell lines and patient-derived xenograft models. Clinically, our analysis reveals a positive correlation between levels of phosphorylated AMPKα (p-AMPKα), phosphorylated Ser123-BAP1 (pSer123-BAP1), and pVHL levels in PDAC, colorectal cancer, and ovarian cancer specimens. Collectively, these findings elucidate a novel mechanism linking dysregulated glucose metabolism to compromised function of the BAP1-pVHL tumor-suppressive axis. Our results suggest that therapeutic strategies designed to activate this pathway may represent a promising approach for treating cancers characterized by downregulated wild-type VHL and aberrant glucose metabolism.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145181838","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epithelial MST1 deficiency promotes pyroptosis and aggravates inflammatory bowel disease via the YAP/p73 signaling pathway. 上皮MST1缺乏通过YAP/p73信号通路促进焦亡并加重炎症性肠病。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-09-26 DOI: 10.1038/s41418-025-01588-3
Jiali Lu, Fei Li, Hailin Wang, Yali Yu, Yuan Yuan, Yukang Zhang, Pule Liu, Qiu Zhao, Min Wu, Mei Ye
{"title":"Epithelial MST1 deficiency promotes pyroptosis and aggravates inflammatory bowel disease via the YAP/p73 signaling pathway.","authors":"Jiali Lu, Fei Li, Hailin Wang, Yali Yu, Yuan Yuan, Yukang Zhang, Pule Liu, Qiu Zhao, Min Wu, Mei Ye","doi":"10.1038/s41418-025-01588-3","DOIUrl":"https://doi.org/10.1038/s41418-025-01588-3","url":null,"abstract":"<p><p>The Hippo pathway has been implicated in the onset and pathogenesis of inflammatory bowel disease (IBD), with Mammalian STE20-like kinase 1 (MST1), a core kinase in this pathway, playing significant roles in inflammation and immune regulation. However, the specific role of MST1 in IBD remains largely undefined. In this study, we observed that MST1 expression was significantly decreased in IBD patients and acute colitis mice. Intestinal epithelial cell-specific MST1 knockout mice exhibited heightened susceptibility to dextran sodium sulfate (DSS)-induced colitis, characterized by severe disruption of intestinal epithelial barrier and markedly increased epithelial cell pyroptosis, thus exacerbating intestinal inflammation. Pharmacological inhibition of caspase-1/GSDMD-mediated pyroptosis ameliorated the detrimental effects of MST1 deficiency in colitis. Consistently, MST1 deficiency exacerbated intestinal barrier disruption and pyroptosis in both in vivo and in vitro models under TNFα-induced inflammation and DNA damage. Mechanistically, MST1 depletion promoted YAP nuclear translocation and enhances its interaction with p73 in intestinal epithelial cells, leading to increased p73 stability and transcriptional activity. This, in turn, facilitated the recruitment of p73 to the caspase-1 promoter, upregulating caspase-1 expression and translating into increased pyroptosis under TNFα-induced inflammatory conditions. Altogether, our findings highlight the critical role of MST1 in maintaining intestinal mucosal barrier homeostasis by regulating epithelial cell pyroptosis via the YAP/p73 signaling pathway. Reduced MST1 expression may correlate with a better response to anti-TNF therapy in IBD patients. Consequently, MST1 could serve as a promising predictive biomarker for anti-TNF therapy responsiveness and a potential therapeutic target for IBD, offering valuable insights for personalized treatment strategies.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145173599","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
USP2 promotes metabolic dysfunction-associated steatotic liver disease progression via stabilization of PPARγ. USP2通过稳定PPARγ促进代谢功能障碍相关的脂肪变性肝病进展。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-09-24 DOI: 10.1038/s41418-025-01589-2
Hao Luo, Chujiao Zhu, Yingying Wang, Yidong Dai, Peng Hao, Haiyan Cai, Wenhui Bai, Zhenge Zhang, Jiale Wan, Youping Zhang, Yun Sun, Ziwei Zhang, Yunzhao Wu, Yuanhui Zhai, Wenxuan Wu, Hu Lei, Hanzhang Xu, Ming He, Yingli Wu
{"title":"USP2 promotes metabolic dysfunction-associated steatotic liver disease progression via stabilization of PPARγ.","authors":"Hao Luo, Chujiao Zhu, Yingying Wang, Yidong Dai, Peng Hao, Haiyan Cai, Wenhui Bai, Zhenge Zhang, Jiale Wan, Youping Zhang, Yun Sun, Ziwei Zhang, Yunzhao Wu, Yuanhui Zhai, Wenxuan Wu, Hu Lei, Hanzhang Xu, Ming He, Yingli Wu","doi":"10.1038/s41418-025-01589-2","DOIUrl":"https://doi.org/10.1038/s41418-025-01589-2","url":null,"abstract":"<p><p>Metabolic dysfunction-associated steatotic liver disease (MASLD) is a leading cause of chronic liver disease worldwide, yet the molecular mechanisms underlying its pathogenesis are not fully understood. Here, we identify the deubiquitinating enzyme Ubiquitin-specific protease 2 (USP2) as a key regulator in hepatic lipid metabolism and MASLD progression. We show that USP2 expression is significantly upregulated in liver tissues from MASLD patients and high-fat diet (HFD)-induced mouse models. Usp2 knockout or pharmacological inhibition alleviates hepatic steatosis and improves systemic metabolic parameters both in vivo and in vitro. Strikingly, hepatocyte-targeted GalNAc-conjugated siRNA against Usp2 markedly attenuates MASLD in mouse models, highlighting therapeutic potential. Mechanistically, USP2 directly interacts with and stabilizes peroxisome proliferator-activated receptor γ (PPARγ) by removing K48-linked ubiquitin chains at lysine 161 within its DNA-binding domain, thereby preventing proteasomal degradation and enhancing its transcriptional activity. This USP2-PPARγ axis promotes hepatic lipid accumulation and drives MASLD progression. Our findings uncover a novel regulatory mechanism in MASLD pathogenesis and suggest that USP2 may represent a promising and druggable therapeutic target for metabolic liver disease.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145136745","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
AARS1-mediated lactylation of H3K18 and STAT1 promotes ferroptosis in diabetic nephropathy. aars1介导的H3K18和STAT1的乳酸化促进糖尿病肾病中的铁下垂。
IF 15.4 1区 生物学
Cell Death and Differentiation Pub Date : 2025-09-23 DOI: 10.1038/s41418-025-01587-4
Jia Hong, Hongjiao Xu, Lang Yu, Zhuang Yu, Xiangyuan Chen, Zhipeng Meng, Jiali Zhu, Jinbao Li, Minmin Zhu
{"title":"AARS1-mediated lactylation of H3K18 and STAT1 promotes ferroptosis in diabetic nephropathy.","authors":"Jia Hong, Hongjiao Xu, Lang Yu, Zhuang Yu, Xiangyuan Chen, Zhipeng Meng, Jiali Zhu, Jinbao Li, Minmin Zhu","doi":"10.1038/s41418-025-01587-4","DOIUrl":"https://doi.org/10.1038/s41418-025-01587-4","url":null,"abstract":"<p><p>Diabetic nephropathy (DN) is the primary cause of end-stage renal disease worldwide. Recent studies have revealed that lactate-mediated histone lactylation, which functions as a novel epigenetic modification, is involved in the occurrence and development of diabetes-related complications. However, little is known about the role of lactyltransferase in DN. Alanyl-tRNA synthetase 1 (AARS1) was identified as a novel lactyltransferase that modulates histone H3-lysine-18 lactylation (H3K18la). In the present study, we determined whether AARS1-mediated H3K18la participates in the pathogenesis of DN. More importantly, we explored the potential mechanism involved. A mouse DN model consisting of both wild-type and alanyl-tRNA synthetase (AARS1) heterozygote (AARS1<sup>+/-</sup>) mice was utilized in this study. Transcriptomic and lipidomic analyses, combined with a variety of molecular biological methodologies, were employed to elucidate the potential mechanism by which AARS1 regulates ferroptosis in DN. Our results indicated that the increases in AARS1 and H3K18la expression were involved in kidney dysfunction and renal cell death via the modulation of ferroptosis in the DN model. Moreover, AARS1 induced lipid peroxidation by increasing fatty acid elongase-5 (ELOVL5) transcription, ultimately contributing to ferroptosis induction. Furthermore, AARS1 interacted with signal transducer and activator of transcription 1 (STAT1) to jointly regulate ELOVL5 transcription. Additionally, treatment with the STAT1-specific inhibitor fludarabine delayed DN progression. In addition, we observed that AARS1 modulated the lactylation of both STAT1 and H3K18 to regulate ELOVL5 transcription, thus triggering ferroptosis. Inhibition of AARS1-induced lactylation via β-alanine attenuated ferroptosis in DN model mice and hyperglycaemic cells. The present study showed that AARS1 induced the lactylation of H3K18 and STAT1 to regulate ELOVL5 transcription, thus triggering ferroptosis in a diabetic nephropathy model.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145129880","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信