首页 > 最新文献

Cell Death and Differentiation最新文献

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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

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.

肺组织糖酵解升高是败血症诱导的急性肺损伤(SI-ALI)的标志,然而糖酵解重编程和乳酸衍生蛋白修饰在损伤上皮细胞中的作用仍然知之甚少。在这项研究中,我们揭示了pdk4驱动的糖酵解重编程促进了SI-ALI期间肺组织中过量的乳酸生成。在机制上,上皮细胞中的AARS1选择性地增强LPCAT2 K375位点的乳酸化修饰,从而抑制STAT1的乙酰化,促进STAT1磷酸化、核易位和SLC7A11的转录抑制。这个级联最终触发上皮细胞铁下垂。药物抑制PDK4可减少乳酸积累和LPCAT2的乳酸化,从而恢复STAT1乙酰化和SLC7A11的表达。此外,在体外和脓毒症小鼠中,AARS1敲低或突变LPCAT2-K375乳酸化位点可挽救stat1介导的SLC7A11抑制,并减轻铁下垂。我们的研究结果显示,PDK4的表达升高是脓毒症期间肺组织乳酸生成增加的一个关键因素,并建立了一个新的LPCAT2-K375/STAT1/SLC7A11轴驱动SI-ALI上皮细胞铁死亡,突出了代谢重编程、翻译后修饰(PTM)和铁死亡之间的串串。靶向PDK4或LPCAT2乳酸化可能为SI-ALI提供治疗潜力。在脓毒症诱导的急性肺损伤(SI-ALI)中,PDK4过度激活驱动上皮细胞过量的乳酸生成,触发AARS1/ hdac9介导的LPCAT2乳酸化。这种修饰抑制STAT1乙酰化,同时增强磷酸化,驱动其核易位和随后的SLC7A11转录下调。由此产生的谷胱甘肽合成缺乏促进铁下垂,加剧SI-ALI的进展。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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
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.
Caspase-8在许多肿瘤中表达上调,尽管它具有典型的凋亡途径,但它维持癌症进展,促进细胞迁移、NF-kB激活和炎症。在这里,我们为Caspase-8在促进癌细胞代谢重布线中的新作用提供了第一个证据。通过对胶质母细胞瘤细胞模型进行转录组学、蛋白质组学和磷酸化蛋白质组学分析,我们发现Caspase-8是一种意想不到的NRF2调节剂。本研究表明,Caspase-8的表达和磷酸化影响NRF2活性和线粒体稳态。从机制上讲,我们证明了酪氨酸380 (Y380)上的src依赖性Caspase-8磷酸化,经常在包括胶质母细胞瘤在内的癌症中报道,维持mTORC1激活,从而促进能量代谢。mTORC1活性导致p62磷酸化,从而允许其依赖于KEAP1蛋白的隔离和构成NRF2信号激活。总的来说,这项工作描述了Caspase-8在调节癌细胞代谢中的一个意想不到的新作用,它将Src、mTORC1和NRF2信号连接在一起。
{"title":"Caspase-8 expression and its Src dependent phosphorylation on Tyrosine 380 triggers NRF2 signaling activation in glioblastoma","authors":"Claudia Cirotti,&nbsp;Claudia Di Girolamo,&nbsp;Irene Taddei,&nbsp;Claudia Contadini,&nbsp;Giorgia Massacci,&nbsp;Francesca Sacco,&nbsp;Donatella Del Bufalo,&nbsp;Illari Salvatori,&nbsp;Cristiana Valle,&nbsp;Daniela Barilà","doi":"10.1038/s41418-025-01542-3","DOIUrl":"10.1038/s41418-025-01542-3","url":null,"abstract":"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.","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"32 12","pages":"2355-2367"},"PeriodicalIF":15.4,"publicationDate":"2025-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.comhttps://www.nature.com/articles/s41418-025-01542-3.pdf","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":"OA","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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

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+ 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+ 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.

肿瘤微环境是一个免疫抑制生态位,通过下调免疫细胞功能或抑制免疫细胞浸润来促进肿瘤生长。人们对其潜在的机制还不是很了解。在这里,我们证明了O-linked N-acetylglucosamine (o - glcnac酰化),一种普遍的蛋白质糖基化形式,通过调节尿苷二磷酸葡萄糖脱氢酶(UGDH)的代谢和非代谢功能,有助于建立免疫抑制生态位。携带o - glcn酰化缺陷UGDH的肿瘤细胞在小鼠中显示出异种移植物肿瘤生长减少和生存率提高。细胞飞行时间(CyTOF)分析显示,UGDH o - glcn酰化与细胞毒性CD8+ T细胞浸润呈负相关。UGDH丝氨酸350上的o - glcn酰化位于udp结合区域内,随后广泛的全原子分子动力学模拟表明,o - glcn酰化增强了UGDH的氢键相互作用和酶活性,从而增强了细胞外基质中透明质酸(HA)的合成。此外,UGDH的o - glcn酰化通过降低趋化因子CXCL10的表达来减少CD8+ T细胞的浸润。具体来说,o - glcn酰化增强了UGDH与KPNA2的相互作用,从而与STAT1竞争,抑制STAT1转位进入细胞核,从而转录下调CXCL10的表达。因此,我们的研究确定了UGDH o - glcn酰化是肿瘤免疫的关键调节因子,并进一步提出了增强免疫治疗的潜在策略。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-04 DOI: 10.1038/s41418-025-01592-7
Siru Zhou, Yulin Li, Chao Wang, Yuhan Zhao, Xiaofeng Zheng

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.

胞质醛缩酶A (ALDOA)在糖酵解中的作用是公认的。然而,ALDOA的细胞质到细胞核的再分布及其核功能尚不清楚。在这里,我们揭示了炎症因子刺激的ALDOA的核功能,通过以泛素化依赖的方式激活NF-κB信号,从而增强胰腺癌的发生。TNF-α触发的ALDOA在Lys200位点的K11-和k29 -连锁泛素化促进了其与RelA/p65的相互作用,促进了进口蛋白β依赖的核易位,通过提高胰腺导管腺癌(pancreatic ductal adencarcinoma, PDAC)中TNF-α的表达,在肿瘤微环境中建立了正反馈调节。USP4是一种负调控因子,可使ALDOA去泛素化。与广泛靶向引起糖酵解损伤的ALDOA不同,特异性消除ALDOA泛素化通过选择性抑制炎症诱导的癌细胞增殖,增强了化疗敏感性和化疗与p65特异性抗炎治疗联合的协同效应。总之,我们揭示了ALDOA促进PDAC致癌的多方面机制,从代谢到基因调控的角度,为对抗癌症提供了潜在的治疗方法。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

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 L-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.

神经活动驱动发育中的大脑血管(BV)形成和能量底物输送,以满足不断上升的代谢需求;然而,潜在的机制仍然知之甚少。在这项研究中,我们将新生小鼠暴露于慢性须刺激(WS),这是一种已知可以增强体感(S1)皮层BV形成的范式。转录组学(RNA-seq)和空间分析(RNA-scope)显示,WS上调皮质神经元中的单羧酸转运蛋白2 (MCT2)和内皮细胞(ECs)中的MCT1。这些变化与皮质乳酸水平升高、星形细胞血管内皮生长因子A (VEGFa)升高和血管生成增强同时发生。功能实验表明,神经元MCT2在介导ws诱导的血管生成和代谢反应中是必不可少的。在机制上,MCT2促进l -乳酸流入皮层,无论有无WS,促进神经元和星形胶质细胞对乳酸的摄取。这进而诱导神经元中MCT2的表达,激活星形胶质细胞中缺氧诱导因子1α (HIF1α)和VEGFa的表达。总之,这些发现揭示了神经元MCT2在调节乳酸通量、信号传导和血管重塑中的作用,从而将神经活动与新生小鼠新皮层的代谢适应和血管发育联系起来。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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":"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

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.

von Hippel-Lindau (VHL)蛋白(pVHL)作为一种有效的肿瘤抑制因子,通过介导多种底物的降解或失活,包括HIFα和Akt。然而,pVHL在许多携带野生型VHL的癌症中经常下调,其潜在机制尚不清楚。异常的葡萄糖代谢是癌症的一个标志,驱动肿瘤进展和治疗抵抗。尽管如此,葡萄糖稳态与pVHL转换和功能之间的联系尚未明确。在这项研究中,我们证明了葡萄糖代谢失调会破坏胰腺导管腺癌(PDAC)、结直肠癌和卵巢癌细胞中pVHL的稳定性。机制上,葡萄糖饥饿、2-脱氧葡萄糖(2-DG)或二甲双胍诱导的能量应激激活amp活化的蛋白激酶(AMPK), AMPK随后磷酸化并激活BAP1, BAP1是一种去泛素酶,其靶向pVHL去泛素化和稳定的特定功能此前尚未被表征。具体来说,AMPKα磷酸化BAP1的S123、S469和S583残基,增强BAP1和pVHL的相互作用,促进pVHL的稳定和肿瘤抑制功能,无论在体内还是体外。相反,通过磷酸化缺陷BAP1突变体(S123A/S469A/S583A)的AMPKα缺失或重构破坏BAP1磷酸化,可以消除BAP1-pVHL的相互作用,导致pVHL稳定性受损,加速癌细胞系和患者来源的异种移植模型的肿瘤进展。临床分析显示,PDAC、结直肠癌和卵巢癌标本中磷酸化AMPKα (p-AMPKα)、磷酸化Ser123-BAP1 (pSer123-BAP1)和pVHL水平呈正相关。总的来说,这些发现阐明了一种将糖代谢失调与BAP1-pVHL肿瘤抑制轴功能受损联系起来的新机制。我们的研究结果表明,旨在激活该途径的治疗策略可能是治疗以下调野生型VHL和异常葡萄糖代谢为特征的癌症的一种有希望的方法。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

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.

Hippo通路与炎症性肠病(IBD)的发病和发病机制有关,该通路的核心激酶哺乳动物ste20样激酶1 (MST1)在炎症和免疫调节中发挥重要作用。然而,MST1在IBD中的具体作用仍未明确。在本研究中,我们观察到MST1在IBD患者和急性结肠炎小鼠中的表达显著降低。肠道上皮细胞特异性MST1基因敲除小鼠对葡聚糖硫酸钠(DSS)诱导的结肠炎的易感性增加,其特征是肠上皮屏障严重破坏,上皮细胞焦亡明显增加,从而加剧肠道炎症。药物抑制caspase-1/ gsdmd介导的焦亡可改善MST1缺乏对结肠炎的有害影响。同样,在tnf α诱导的炎症和DNA损伤的体内和体外模型中,MST1缺乏都加剧了肠屏障破坏和焦亡。在机制上,MST1缺失促进肠上皮细胞中YAP核易位并增强其与p73的相互作用,导致p73稳定性和转录活性增加。这反过来又促进了p73向caspase-1启动子的募集,上调caspase-1的表达,并在tnf α诱导的炎症条件下转化为增加的焦亡。总之,我们的研究结果强调了MST1通过YAP/p73信号通路调节上皮细胞焦亡在维持肠粘膜屏障稳态中的关键作用。减少MST1表达可能与IBD患者对抗tnf治疗的更好反应相关。因此,MST1可以作为抗tnf治疗反应性的有希望的预测性生物标志物和IBD的潜在治疗靶点,为个性化治疗策略提供有价值的见解。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

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.

代谢功能障碍相关脂肪变性肝病(MASLD)是世界范围内慢性肝病的主要病因,但其发病机制的分子机制尚不完全清楚。在这里,我们发现去泛素化酶泛素特异性蛋白酶2 (USP2)是肝脏脂质代谢和MASLD进展的关键调节因子。我们发现,在MASLD患者和高脂肪饮食(HFD)诱导的小鼠模型的肝组织中,USP2的表达显著上调。Usp2基因敲除或药物抑制可减轻肝脂肪变性,改善体内和体外的全身代谢参数。引人注目的是,针对Usp2的肝细胞靶向galnac偶联siRNA在小鼠模型中显着减弱了MASLD,突出了治疗潜力。在机制上,USP2通过去除dna结合域内赖氨酸161上的k48连接的泛素链,直接与过氧化物酶体增殖体激活受体γ (PPARγ)相互作用并稳定PPARγ,从而阻止蛋白酶体降解并增强其转录活性。USP2-PPARγ轴促进肝脏脂质积累并驱动MASLD进展。我们的研究结果揭示了MASLD发病机制的一种新的调控机制,并表明USP2可能是代谢性肝病的一个有希望的可药物治疗靶点。
{"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 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY 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

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+/-) 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.

糖尿病肾病(DN)是终末期肾脏疾病的主要原因。最近的研究表明,乳酸介导的组蛋白乳酸化作为一种新的表观遗传修饰,参与了糖尿病相关并发症的发生和发展。然而,对乳酸转移酶在DN中的作用知之甚少。Alanyl-tRNA合成酶1 (AARS1)是一种调节组蛋白h3 -赖氨酸-18乳酸化(H3K18la)的新型乳酸转移酶。在本研究中,我们确定了aars1介导的H3K18la是否参与了DN的发病机制。更重要的是,我们探索了潜在的机制。本研究采用野生型和alanyl-tRNA合成酶(AARS1)杂合子(AARS1+/-)小鼠组成的小鼠DN模型。转录组学和脂质组学分析结合多种分子生物学方法,阐明了AARS1调控DN中铁下垂的潜在机制。我们的研究结果表明,在DN模型中,AARS1和H3K18la表达的增加通过对铁下沉的调节参与肾功能障碍和肾细胞死亡。此外,AARS1通过增加脂肪酸延长酶-5 (ELOVL5)转录诱导脂质过氧化,最终导致铁下垂。此外,AARS1与信号换能器和转录激活器1 (STAT1)相互作用,共同调控ELOVL5的转录。此外,用stat1特异性抑制剂氟达拉滨治疗可以延缓DN的进展。此外,我们观察到AARS1通过调节STAT1和H3K18的乳酸化来调节ELOVL5的转录,从而引发铁凋亡。通过β-丙氨酸减轻DN模型小鼠和高血糖细胞的铁下垂抑制aars1诱导的乳酸化。本研究表明,在糖尿病肾病模型中,AARS1诱导H3K18和STAT1的乳酸化,从而调控ELOVL5的转录,从而引发铁凋亡。
{"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
期刊
Cell Death and Differentiation
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1