首页 > 最新文献

Cell Death and Differentiation最新文献

英文 中文
Induction of ferroptosis in prostate cancer by CCDC719-13 via TRIM21-mediated ubiquitination of SLC7A11. 通过trim21介导的SLC7A11泛素化,CCDC719-13诱导前列腺癌铁凋亡。
IF 15.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-22 DOI: 10.1038/s41418-025-01580-x
Bisheng Cheng, Qiong Wang, Zean Li, Tianlong Luo, JunJia Xie, Sandeep Singh, Yong Luo, Xu Gao, Hui Li, Zongwei Wang, Peng Wu, Hai Huang

Prostate cancer is one of the most prevalent malignancies in men, with increasing incidence and mortality largely attributed to treatment resistance and metastasis. The effectiveness of current therapies for advanced cases is hindered by intricate genetic and microenvironmental factors, emphasizing the urgent need for novel therapeutic targets. Chimeric RNAs have emerged as promising biomarkers in cancer research, among which CCDC719-13, a circular chimeric RNA, is frequently identified in prostate cancer. Our study reveals that CCDC719-13 expression is markedly reduced in advanced and recurrent prostate cancer, where its low levels serve as an independent predictor of poor prognosis. Functional experiments demonstrate that CCDC719-13 overexpression inhibits cell proliferation, induces apoptosis, and suppresses tumor growth in vivo, whereas its knockdown reverses these effects. Mechanistically, CCDC719-13 encodes a novel protein, CCDC7241aa, which triggers ferroptosis by interacting with SLC7A11 and facilitating its TRIM21-mediated ubiquitination and degradation. Notably, treatment with recombinant CCDC7241aa effectively suppresses tumor growth in patient-derived xenograft models without toxicity and enhances the efficacy of docetaxel and enzalutamide in vitro. These findings establish CCDC719-13 as a significant prognostic marker and potential therapeutic target in prostate cancer, with the recombinant CCDC7241aa protein offering promise for combination therapies in advanced cases.

前列腺癌是男性最常见的恶性肿瘤之一,其发病率和死亡率的增加主要归因于治疗耐药性和转移。目前治疗晚期病例的有效性受到复杂的遗传和微环境因素的阻碍,强调迫切需要新的治疗靶点。嵌合RNA已成为癌症研究中很有前景的生物标志物,其中环状嵌合RNA CCDC719-13在前列腺癌中经常被发现。我们的研究表明,CCDC719-13在晚期和复发性前列腺癌中表达显著降低,其低水平可作为预后不良的独立预测因子。功能实验表明,CCDC719-13过表达可抑制细胞增殖,诱导细胞凋亡,抑制肿瘤生长,而其敲低可逆转这些作用。在机制上,CCDC719-13编码一种新的蛋白CCDC7241aa,该蛋白通过与SLC7A11相互作用并促进其trim21介导的泛素化和降解,从而引发铁死亡。值得注意的是,用重组CCDC7241aa治疗可有效抑制患者来源的异种移植模型中的肿瘤生长,且无毒性,并增强了多西他赛和恩杂鲁胺的体外疗效。这些研究结果表明,CCDC719-13是前列腺癌重要的预后标志物和潜在的治疗靶点,重组CCDC7241aa蛋白为晚期病例的联合治疗提供了希望。
{"title":"Induction of ferroptosis in prostate cancer by CCDC7<sub>19-13</sub> via TRIM21-mediated ubiquitination of SLC7A11.","authors":"Bisheng Cheng, Qiong Wang, Zean Li, Tianlong Luo, JunJia Xie, Sandeep Singh, Yong Luo, Xu Gao, Hui Li, Zongwei Wang, Peng Wu, Hai Huang","doi":"10.1038/s41418-025-01580-x","DOIUrl":"10.1038/s41418-025-01580-x","url":null,"abstract":"<p><p>Prostate cancer is one of the most prevalent malignancies in men, with increasing incidence and mortality largely attributed to treatment resistance and metastasis. The effectiveness of current therapies for advanced cases is hindered by intricate genetic and microenvironmental factors, emphasizing the urgent need for novel therapeutic targets. Chimeric RNAs have emerged as promising biomarkers in cancer research, among which CCDC7<sub>19-13</sub>, a circular chimeric RNA, is frequently identified in prostate cancer. Our study reveals that CCDC7<sub>19-13</sub> expression is markedly reduced in advanced and recurrent prostate cancer, where its low levels serve as an independent predictor of poor prognosis. Functional experiments demonstrate that CCDC7<sub>19-13</sub> overexpression inhibits cell proliferation, induces apoptosis, and suppresses tumor growth in vivo, whereas its knockdown reverses these effects. Mechanistically, CCDC7<sub>19-13</sub> encodes a novel protein, CCDC7<sub>241aa</sub>, which triggers ferroptosis by interacting with SLC7A11 and facilitating its TRIM21-mediated ubiquitination and degradation. Notably, treatment with recombinant CCDC7<sub>241aa</sub> effectively suppresses tumor growth in patient-derived xenograft models without toxicity and enhances the efficacy of docetaxel and enzalutamide in vitro. These findings establish CCDC7<sub>19-13</sub> as a significant prognostic marker and potential therapeutic target in prostate cancer, with the recombinant CCDC7<sub>241aa</sub> protein offering promise for combination therapies in advanced cases.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145124117","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
A novel lncRNA, lncMCL1, modulates neural pyroptosis associated with epilepsy via stabilizing DDX3X. 一种新的lncRNA lncMCL1通过稳定DDX3X调节与癫痫相关的神经焦亡。
IF 15.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-22 DOI: 10.1038/s41418-025-01584-7
Huizhi Wang, Liuliu Wu, Chong Liu, Xueming Zhao, Luhao Cui, Jianing Gao, Chaonan Zhang, Tingting Du, Lin Shi, Yuchen Ji, Yilei Xiao, Jianguo Zhang, Wenjun Tu, Fangang Meng, Chunlei Han

Pyroptosis is strongly associated with refractory epilepsy. However, the underlying mechanisms remain poorly understood. Increasing evidence has shown that long noncoding RNAs (lncRNAs) participate in various neurological disorder processes by regulating programmed cell death. In this study, we identified a novel lncRNA, lncMCL1, by high-throughput screening, which suppresses NLRP3 inflammasome-dependent neural pyroptosis in epilepsy. We demonstrated that lncMCL1 is aberrantly underexpressed in the hippocampus and cortex of epilepsy patients, a phenomenon that was validated in various mouse and rat epilepsy models. Through CRISPR/Cas9, siRNA, and viral manipulation, gain- and loss-of-function experiments confirmed that lncMCL1 inhibits neuronal pyroptosis in vivo and in vitro and exerts antiepileptic effects. Mechanistically, lncMCL1 acts as a scaffold to modulate DDX3X protein stabilization by enhancing NEDD4-mediated DDX3X K48 ubiquitination, thereby inhibiting neural pyroptosis through the suppression of NLRP3 inflammasome signalling. Additionally, IL-18/IL-1β, downstream cytokines of pyroptosis, inhibit lncMCL1 expression through the activation of a shared pathway, the STAT3 pathway, forming a feedback loop. Our findings identify lncMCL1 as a critical regulator of neural cell pyroptosis and a promising therapeutic target for refractory epilepsy.

焦下垂与难治性癫痫密切相关。然而,潜在的机制仍然知之甚少。越来越多的证据表明,长链非编码rna (lncRNAs)通过调节程序性细胞死亡参与各种神经系统疾病过程。在这项研究中,我们通过高通量筛选发现了一种新的lncRNA lncMCL1,它可以抑制癫痫患者NLRP3炎症小体依赖性神经焦亡。我们证明lncMCL1在癫痫患者的海马和皮质中异常低表达,这一现象在各种小鼠和大鼠癫痫模型中得到了验证。通过CRISPR/Cas9、siRNA和病毒操作,功能获得和功能丧失实验证实lncMCL1在体内和体外抑制神经元焦亡并具有抗癫痫作用。机制上,lncMCL1作为支架通过增强nedd4介导的DDX3X K48泛素化来调节DDX3X蛋白的稳定性,从而通过抑制NLRP3炎症小体信号传导来抑制神经焦亡。此外,焦亡的下游细胞因子IL-18/IL-1β通过激活一个共享通路STAT3通路抑制lncMCL1的表达,形成一个反馈回路。我们的研究结果确定lncMCL1是神经细胞焦亡的关键调节因子,也是难治性癫痫的一个有希望的治疗靶点。
{"title":"A novel lncRNA, lncMCL1, modulates neural pyroptosis associated with epilepsy via stabilizing DDX3X.","authors":"Huizhi Wang, Liuliu Wu, Chong Liu, Xueming Zhao, Luhao Cui, Jianing Gao, Chaonan Zhang, Tingting Du, Lin Shi, Yuchen Ji, Yilei Xiao, Jianguo Zhang, Wenjun Tu, Fangang Meng, Chunlei Han","doi":"10.1038/s41418-025-01584-7","DOIUrl":"https://doi.org/10.1038/s41418-025-01584-7","url":null,"abstract":"<p><p>Pyroptosis is strongly associated with refractory epilepsy. However, the underlying mechanisms remain poorly understood. Increasing evidence has shown that long noncoding RNAs (lncRNAs) participate in various neurological disorder processes by regulating programmed cell death. In this study, we identified a novel lncRNA, lncMCL1, by high-throughput screening, which suppresses NLRP3 inflammasome-dependent neural pyroptosis in epilepsy. We demonstrated that lncMCL1 is aberrantly underexpressed in the hippocampus and cortex of epilepsy patients, a phenomenon that was validated in various mouse and rat epilepsy models. Through CRISPR/Cas9, siRNA, and viral manipulation, gain- and loss-of-function experiments confirmed that lncMCL1 inhibits neuronal pyroptosis in vivo and in vitro and exerts antiepileptic effects. Mechanistically, lncMCL1 acts as a scaffold to modulate DDX3X protein stabilization by enhancing NEDD4-mediated DDX3X K48 ubiquitination, thereby inhibiting neural pyroptosis through the suppression of NLRP3 inflammasome signalling. Additionally, IL-18/IL-1β, downstream cytokines of pyroptosis, inhibit lncMCL1 expression through the activation of a shared pathway, the STAT3 pathway, forming a feedback loop. Our findings identify lncMCL1 as a critical regulator of neural cell pyroptosis and a promising therapeutic target for refractory epilepsy.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145124034","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
Male specific conserved LncRNA TSCL1 regulated target mRNA translation by interaction with PIWIL1. 男性特异性保守LncRNA TSCL1通过与PIWIL1相互作用调控靶mRNA翻译。
IF 15.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-20 DOI: 10.1038/s41418-025-01583-8
Shuai Lu, Yang Li, Chenmeijie Li, Zhongyu Zou, Xiaoxi Xu, Shijie Zhu, Beibei Yang, Gaoming Tang, Haoran Chen, Yuchen Wang, Feng Li, Na Qin, Cheng Wang, Hongbing Shen, Zhibin Hu, Yayun Gu

Long non-coding RNAs (lncRNAs) play crucial roles in diverse mammalian physiological processes, yet their functions in spermatogenesis remain largely underexplored. Here, we identify a unique class of conserved haploid spermatid-associated lncRNAs (cHS-LncRNAs) defined by sequence conservation, testis-restricted expression, and elevated levels in haploid spermatids. Among these, testis-specific conserved lncRNA 1 (Tscl1) is the most highly expressed in round spermatids. Tscl1-null male mice exhibit reduced sperm motility, disorganized mitochondrial sheaths, abnormal fatty acid metabolism, and complete infertility. Mechanistically, Tscl1 directly binds PIWIL1 and HuR via its 5' stem-loop and multiple AU-rich elements, respectively. This interaction promotes assembly of a PIWIL1/eIF3f/HuR/eIF4G3 complex that enhances translation of fatty-acid-metabolism-related mRNAs within the chromatoid body. Notably, TSCL1 variants disrupting the PIWIL1-binding region are significantly enriched in patients with non-obstructive azoospermia (NOA) compared to fertile controls. Collectively, our findings uncover a critical role for Tscl1 in modulating translation during spermiogenesis and implicate TSCL1 as a potential pathogenic locus in human male infertility.

长链非编码rna (lncRNAs)在多种哺乳动物生理过程中发挥着至关重要的作用,但其在精子发生中的功能仍未得到充分研究。在这里,我们鉴定了一类独特的保守的单倍体精子相关lncRNAs (cHS-LncRNAs),这些lncRNAs由序列保守、睾丸限制性表达和单倍体精子中水平升高定义。其中,睾丸特异性保守lncRNA 1 (Tscl1)在圆形精子中表达量最高。tscl1缺失的雄性小鼠表现为精子活力降低、线粒体鞘紊乱、脂肪酸代谢异常和完全不育。在机制上,Tscl1分别通过其5'茎环和多个富au元件直接结合PIWIL1和HuR。这种相互作用促进PIWIL1/eIF3f/HuR/eIF4G3复合物的组装,从而增强染色质体内脂肪酸代谢相关mrna的翻译。值得注意的是,与生育对照组相比,破坏piwil1结合区域的tsc1变异在非阻塞性无精子症(NOA)患者中显著富集。总之,我们的研究结果揭示了Tscl1在精子发生过程中调节翻译的关键作用,并暗示Tscl1是人类男性不育的潜在致病位点。
{"title":"Male specific conserved LncRNA TSCL1 regulated target mRNA translation by interaction with PIWIL1.","authors":"Shuai Lu, Yang Li, Chenmeijie Li, Zhongyu Zou, Xiaoxi Xu, Shijie Zhu, Beibei Yang, Gaoming Tang, Haoran Chen, Yuchen Wang, Feng Li, Na Qin, Cheng Wang, Hongbing Shen, Zhibin Hu, Yayun Gu","doi":"10.1038/s41418-025-01583-8","DOIUrl":"https://doi.org/10.1038/s41418-025-01583-8","url":null,"abstract":"<p><p>Long non-coding RNAs (lncRNAs) play crucial roles in diverse mammalian physiological processes, yet their functions in spermatogenesis remain largely underexplored. Here, we identify a unique class of conserved haploid spermatid-associated lncRNAs (cHS-LncRNAs) defined by sequence conservation, testis-restricted expression, and elevated levels in haploid spermatids. Among these, testis-specific conserved lncRNA 1 (Tscl1) is the most highly expressed in round spermatids. Tscl1-null male mice exhibit reduced sperm motility, disorganized mitochondrial sheaths, abnormal fatty acid metabolism, and complete infertility. Mechanistically, Tscl1 directly binds PIWIL1 and HuR via its 5' stem-loop and multiple AU-rich elements, respectively. This interaction promotes assembly of a PIWIL1/eIF3f/HuR/eIF4G3 complex that enhances translation of fatty-acid-metabolism-related mRNAs within the chromatoid body. Notably, TSCL1 variants disrupting the PIWIL1-binding region are significantly enriched in patients with non-obstructive azoospermia (NOA) compared to fertile controls. Collectively, our findings uncover a critical role for Tscl1 in modulating translation during spermiogenesis and implicate TSCL1 as a potential pathogenic locus in human male infertility.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145102615","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
TRIM24-mediated K27-linked ubiquitination of ULK1 alleviates energy stress-induced autophagy and promote prostate cancer growth in the context of SPOP mutation. trim24介导的k27关联的ULK1泛素化减轻能量应激诱导的自噬,促进SPOP突变背景下前列腺癌的生长。
IF 15.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-20 DOI: 10.1038/s41418-025-01582-9
Shimin Chen, Jichun Lin, Zhan Yang, Yuanjing Wang, Qiang Wang, Dong Wang, Yue Qu, Qian Lin, Jia Liu, Shi Yan, Zixin Wang, Xueyu Qian, Yutian Xiao, Xue Li, Yinuo Chen, Wenshuo Fang, Jiaojiao Zhao, Zhimin Lu, He Ren, Yasheng Zhu, Leina Ma

SPOP, the most frequently mutated gene in prostate cancer, has been implicated in the aberrant activation of stress granules, presenting significant challenges in disease management. However, the mechanistic link between SPOP mutations and cellular energy stress remains inadequately explored. In this study, we demonstrate that ULK1 expression is positively correlated with both loss-of-function mutations in SPOP and the upregulation of the E3 ubiquitin ligase TRIM24 in human prostate cancer specimens. Mechanistically, SPOP mutations induce the upregulation of TRIM24, which subsequently binds to ULK1 and catalyzes its non-degradative K27-linked polyubiquitylation. This post-translational modification enhances the stability of ULK1, facilitating cellular adaptation to energy stress and consequently promoting prostate cancer progression. Notably, pharmacological inhibition of TRIM24 using TRIM24-PROTAC (proteolysis-targeting chimera) effectively suppressed tumor growth in mice bearing SPOP-mutant prostate cancer cells. Collectively, these findings elucidate a pivotal role of SPOP mutations in modulating energy stress responses via TRIM24-mediated ULK1 ubiquitylation and underscore the therapeutic potential of targeting TRIM24 in SPOP-mutant prostate cancers.

SPOP是前列腺癌中最常见的突变基因,与应激颗粒的异常激活有关,对疾病管理提出了重大挑战。然而,SPOP突变与细胞能量应激之间的机制联系仍未得到充分探讨。在这项研究中,我们证明了ULK1的表达与人类前列腺癌标本中SPOP的功能缺失突变和E3泛素连接酶TRIM24的上调呈正相关。从机制上讲,SPOP突变诱导TRIM24上调,TRIM24随后与ULK1结合并催化其不可降解的k27连接的多泛素化。这种翻译后修饰增强了ULK1的稳定性,促进细胞对能量应激的适应,从而促进前列腺癌的进展。值得注意的是,使用TRIM24- protac(蛋白水解靶向嵌合体)对TRIM24进行药理学抑制可以有效抑制携带spop突变前列腺癌细胞的小鼠的肿瘤生长。总之,这些发现阐明了SPOP突变在通过TRIM24介导的ULK1泛素化调节能量应激反应中的关键作用,并强调了靶向TRIM24治疗SPOP突变前列腺癌的潜力。
{"title":"TRIM24-mediated K27-linked ubiquitination of ULK1 alleviates energy stress-induced autophagy and promote prostate cancer growth in the context of SPOP mutation.","authors":"Shimin Chen, Jichun Lin, Zhan Yang, Yuanjing Wang, Qiang Wang, Dong Wang, Yue Qu, Qian Lin, Jia Liu, Shi Yan, Zixin Wang, Xueyu Qian, Yutian Xiao, Xue Li, Yinuo Chen, Wenshuo Fang, Jiaojiao Zhao, Zhimin Lu, He Ren, Yasheng Zhu, Leina Ma","doi":"10.1038/s41418-025-01582-9","DOIUrl":"https://doi.org/10.1038/s41418-025-01582-9","url":null,"abstract":"<p><p>SPOP, the most frequently mutated gene in prostate cancer, has been implicated in the aberrant activation of stress granules, presenting significant challenges in disease management. However, the mechanistic link between SPOP mutations and cellular energy stress remains inadequately explored. In this study, we demonstrate that ULK1 expression is positively correlated with both loss-of-function mutations in SPOP and the upregulation of the E3 ubiquitin ligase TRIM24 in human prostate cancer specimens. Mechanistically, SPOP mutations induce the upregulation of TRIM24, which subsequently binds to ULK1 and catalyzes its non-degradative K27-linked polyubiquitylation. This post-translational modification enhances the stability of ULK1, facilitating cellular adaptation to energy stress and consequently promoting prostate cancer progression. Notably, pharmacological inhibition of TRIM24 using TRIM24-PROTAC (proteolysis-targeting chimera) effectively suppressed tumor growth in mice bearing SPOP-mutant prostate cancer cells. Collectively, these findings elucidate a pivotal role of SPOP mutations in modulating energy stress responses via TRIM24-mediated ULK1 ubiquitylation and underscore the therapeutic potential of targeting TRIM24 in SPOP-mutant prostate cancers.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":15.4,"publicationDate":"2025-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145102567","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
NAT10 regulates heart development and function by maintaining the expression of genes related to fatty acid β-oxidation and heart contraction NAT10通过维持脂肪酸β-氧化和心脏收缩相关基因的表达来调节心脏发育和功能
IF 12.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-13 DOI: 10.1038/s41418-025-01577-6
Lei Shi, Meiwei Zhang, Hao Yang, Xinzhi Li, Siyi He, Yanshuo Chu, Minghui Gao, Zhiguo Zhang, Joe Z. Zhang, Zhuo Li, Zheng Chen

Energy metabolism is crucial for heart development and function, and dysregulation of this process can lead to heart failure. However, the molecular mechanisms underlying these processes, particularly the role of RNA-binding proteins (RBPs)-mediated posttranscriptional regulation, remain largely unclear. We identified N-acetyltransferase 10 (NAT10) as a key regulator of heart function and cardiac diseases. NAT10 is crucial for heart development, and its dysregulation is associated with heart failure. Cardiac-specific deletion of Nat10 leads to dilated cardiomyopathy, heart failure, and postnatal death by downregulating genes related to fatty acid β-oxidation and heart contraction. Adult-onset knockout Nat10 also results in dilated cardiomyopathy and heart failure. NAT10-deficient hiPSC-CMs also showed impaired calcium transients during contraction. Restoration of NAT10(WT) and NAT10(G641E) (an N-acetyltransferase-inactive mutation), but not NAT10(K290A) (a loss-of-RNA-binding activity mutation), fully rescues the dilated cardiomyopathy, heart failure, and postnatal death phenotypes in Nat10-CKO mice by restoring expression of genes involved in fatty acid β-oxidation and heart contraction. The RNA-binding activity of NAT10 is essential for maintaining the expression of these genes. These findings demonstrate that NAT10 plays a critical role in heart development and function by maintaining the expression of genes related to fatty acid β-oxidation and heart contraction, highlighting its importance in maintaining heart health.

能量代谢对心脏发育和功能至关重要,这一过程的失调会导致心力衰竭。然而,这些过程的分子机制,特别是rna结合蛋白(rbp)介导的转录后调控的作用,在很大程度上仍然不清楚。我们发现n -乙酰转移酶10 (NAT10)是心脏功能和心脏疾病的关键调节因子。NAT10对心脏发育至关重要,其失调与心力衰竭有关。心脏特异性缺失Nat10通过下调脂肪酸β-氧化和心脏收缩相关基因导致扩张性心肌病、心力衰竭和产后死亡。成人发病的Nat10基因敲除也会导致扩张性心肌病和心力衰竭。缺乏nat10的hiPSC-CMs在收缩期间也表现出钙瞬态受损。恢复NAT10(WT)和NAT10(G641E)(一种n -乙酰转移酶失活突变),而不是NAT10(K290A)(一种rna结合活性缺失突变),通过恢复脂肪酸β氧化和心脏收缩相关基因的表达,完全挽救了NAT10 - cko小鼠的扩张型心肌病、心力衰竭和出生后死亡表型。NAT10的rna结合活性对于维持这些基因的表达至关重要。这些发现表明,NAT10通过维持脂肪酸β-氧化和心脏收缩相关基因的表达,在心脏发育和功能中起着关键作用,突出了其在维持心脏健康中的重要性。
{"title":"NAT10 regulates heart development and function by maintaining the expression of genes related to fatty acid β-oxidation and heart contraction","authors":"Lei Shi, Meiwei Zhang, Hao Yang, Xinzhi Li, Siyi He, Yanshuo Chu, Minghui Gao, Zhiguo Zhang, Joe Z. Zhang, Zhuo Li, Zheng Chen","doi":"10.1038/s41418-025-01577-6","DOIUrl":"https://doi.org/10.1038/s41418-025-01577-6","url":null,"abstract":"<p>Energy metabolism is crucial for heart development and function, and dysregulation of this process can lead to heart failure. However, the molecular mechanisms underlying these processes, particularly the role of RNA-binding proteins (RBPs)-mediated posttranscriptional regulation, remain largely unclear. We identified N-acetyltransferase 10 (NAT10) as a key regulator of heart function and cardiac diseases. NAT10 is crucial for heart development, and its dysregulation is associated with heart failure. Cardiac-specific deletion of <i>Nat10</i> leads to dilated cardiomyopathy, heart failure, and postnatal death by downregulating genes related to fatty acid β-oxidation and heart contraction. Adult-onset knockout <i>Nat10</i> also results in dilated cardiomyopathy and heart failure. <i>NAT10</i>-deficient hiPSC-CMs also showed impaired calcium transients during contraction. Restoration of NAT10(WT) and NAT10(G641E) (an N-acetyltransferase-inactive mutation), but not NAT10(K290A) (a loss-of-RNA-binding activity mutation), fully rescues the dilated cardiomyopathy, heart failure, and postnatal death phenotypes in <i>Nat10</i>-CKO mice by restoring expression of genes involved in fatty acid β-oxidation and heart contraction. The RNA-binding activity of NAT10 is essential for maintaining the expression of these genes. These findings demonstrate that NAT10 plays a critical role in heart development and function by maintaining the expression of genes related to fatty acid β-oxidation and heart contraction, highlighting its importance in maintaining heart health.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"50 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2025-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145043419","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
NLRP3 autophagic degradation disruption in melanocytes contributes to vitiligo development 黑色素细胞NLRP3自噬降解破坏有助于白癜风的发展
IF 12.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-11 DOI: 10.1038/s41418-025-01578-5
Ke Zeng, Yuqi Zhu, Zhongxin Han, Siyi Xiong, Yan Zhao, Zilong Xiao, Yingchao Xie, Shiyu Jin, Tingru Dong, Lan Lan, Weiwei Liu, Yongzhong Du, Cuiping Guan, Xiao Yu, Xiuzu Song

NLRP3 functions as a critical intracellular danger sensor for inflammasome activation, playing a crucial role in autoimmune diseases. Vitiligo progression has been linked to NLRP3, yet its specific involvement in melanocytes of vitiligo remains poorly understood. In this study, we demonstrate that NLRP3 expression is significantly upregulated in the melanocytes of vitiligo patients and melanoma-Treg-induced vitiligo mouse model. Genetic knockout of NLRP3 effectively alleviates vitiligo progression in these mice. Our mechanistic investigations reveal that the downregulation of the E3 ligase β-TrCP1 in vitiligo melanocytes decreases K27-linked ubiquitination levels of NLRP3, which in turn weakens its interaction with the autophagy receptor NDP52. This disruption impairs the selective autophagic degradation of NLRP3, leading to hyperactivation of inflammation and pyroptosis in melanocytes, thereby accelerating vitiligo pathogenesis. Notably, melanocyte-specific knockdown of NLRP3 using lysine-proline-valine (KPV)-modified deformable liposomes (KPV-Lipos) carrying Nlrp3 shRNA significantly alleviates vitiligo development. This study elucidates the mechanism by which autophagy dysfunction mediated excessive NLRP3 inflammasome activation in melanocytes contributes to vitiligo pathogenesis, highlighting potential therapeutic strategies targeting these pathways for the treatment of vitiligo and other pigment-related skin diseases.

Overview of disrupted NLRP3 autophagic degradation in vitiligo melanocytes. In healthy melanocytes, NLRP3 expression is upregulated when subjected to oxidative stress, along with an increase in the E3 ligase β-TrCP1, which enhances the K27-linked ubiquitination of NLRP3 and further strengthens its binding to the autophagy receptor protein NDP52, thus effectively suppressing the excessive inflammatory response. Whereas in the melanocytes of vitiligo patients, decreased expression of β-TrCP1 leads to downregulation of K27-linked ubiquitination in NLRP3, thus inhibiting its autophagic degradation. The persistent activation of NLRP3 in vitiligo melanocytes promotes the cleavage of pro-IL-1β and GSDMD. GSDMD-N subsequently forms pores on the cell membrane, which causes the release of IL-1β and results in melanocyte pyroptosis. In our study, we utilize KPV-Lipos with Nlrp3 shRNA to precisely knockdown NLRP3 expression in melanocytes and effectively alleviate vitiligo development, which provide a potentially promising strategy for the treatment of vitiligo. MC, melanocytes.

NLRP3作为炎症小体激活的关键细胞内危险传感器,在自身免疫性疾病中发挥重要作用。白癜风的进展与NLRP3有关,但其在白癜风黑色素细胞中的具体作用仍知之甚少。在本研究中,我们证明了NLRP3在白癜风患者和黑色素瘤- treg诱导的白癜风小鼠模型中黑色素细胞的表达显著上调。基因敲除NLRP3可有效缓解这些小鼠的白癜风进展。我们的机制研究表明,白癜风黑素细胞中E3连接酶β-TrCP1的下调会降低NLRP3的k27相关泛素化水平,从而削弱其与自噬受体NDP52的相互作用。这种破坏破坏了NLRP3的选择性自噬降解,导致黑素细胞炎症和焦亡的过度激活,从而加速白癜风的发病。值得注意的是,使用携带NLRP3 shRNA的赖氨酸-脯氨酸-缬氨酸(KPV)修饰的可变形脂质体(KPV- lipos)对NLRP3进行黑素细胞特异性敲除可显著缓解白癜风的发展。本研究阐明了自噬功能障碍介导的黑素细胞NLRP3炎性体过度激活参与白癜风发病的机制,并强调了针对这些途径治疗白癜风和其他色素相关皮肤病的潜在治疗策略。白癜风黑素细胞中NLRP3自噬降解中断的综述。在健康黑素细胞中,NLRP3在氧化应激下表达上调,同时E3连接酶β-TrCP1增加,增强了NLRP3的k27连锁泛素化,进一步加强了其与自噬受体蛋白NDP52的结合,从而有效抑制过度炎症反应。而在白癜风患者的黑色素细胞中,β-TrCP1的表达降低导致NLRP3中k27相关泛素化的下调,从而抑制其自噬降解。白癜风黑色素细胞中NLRP3的持续激活促进了前il -1β和GSDMD的分裂。GSDMD-N随后在细胞膜上形成孔隙,导致IL-1β的释放并导致黑素细胞焦亡。在我们的研究中,我们利用KPV-Lipos结合Nlrp3 shRNA,精确地敲低黑色素细胞中Nlrp3的表达,有效地缓解白癜风的发展,为白癜风的治疗提供了一个潜在的有前景的策略。MC,黑色素细胞。
{"title":"NLRP3 autophagic degradation disruption in melanocytes contributes to vitiligo development","authors":"Ke Zeng, Yuqi Zhu, Zhongxin Han, Siyi Xiong, Yan Zhao, Zilong Xiao, Yingchao Xie, Shiyu Jin, Tingru Dong, Lan Lan, Weiwei Liu, Yongzhong Du, Cuiping Guan, Xiao Yu, Xiuzu Song","doi":"10.1038/s41418-025-01578-5","DOIUrl":"https://doi.org/10.1038/s41418-025-01578-5","url":null,"abstract":"<p>NLRP3 functions as a critical intracellular danger sensor for inflammasome activation, playing a crucial role in autoimmune diseases. Vitiligo progression has been linked to NLRP3, yet its specific involvement in melanocytes of vitiligo remains poorly understood. In this study, we demonstrate that NLRP3 expression is significantly upregulated in the melanocytes of vitiligo patients and melanoma-Treg-induced vitiligo mouse model. Genetic knockout of NLRP3 effectively alleviates vitiligo progression in these mice. Our mechanistic investigations reveal that the downregulation of the E3 ligase β-TrCP1 in vitiligo melanocytes decreases K27-linked ubiquitination levels of NLRP3, which in turn weakens its interaction with the autophagy receptor NDP52. This disruption impairs the selective autophagic degradation of NLRP3, leading to hyperactivation of inflammation and pyroptosis in melanocytes, thereby accelerating vitiligo pathogenesis. Notably, melanocyte-specific knockdown of NLRP3 using lysine-proline-valine (KPV)-modified deformable liposomes (KPV-Lipos) carrying <i>Nlrp3</i> shRNA significantly alleviates vitiligo development. This study elucidates the mechanism by which autophagy dysfunction mediated excessive NLRP3 inflammasome activation in melanocytes contributes to vitiligo pathogenesis, highlighting potential therapeutic strategies targeting these pathways for the treatment of vitiligo and other pigment-related skin diseases.</p><figure><p><b>Overview of disrupted NLRP3 autophagic degradation in vitiligo melanocytes</b>. In healthy melanocytes, NLRP3 expression is upregulated when subjected to oxidative stress, along with an increase in the E3 ligase β-TrCP1, which enhances the K27-linked ubiquitination of NLRP3 and further strengthens its binding to the autophagy receptor protein NDP52, thus effectively suppressing the excessive inflammatory response. Whereas in the melanocytes of vitiligo patients, decreased expression of β-TrCP1 leads to downregulation of K27-linked ubiquitination in NLRP3, thus inhibiting its autophagic degradation. The persistent activation of NLRP3 in vitiligo melanocytes promotes the cleavage of pro-IL-1β and GSDMD. GSDMD-N subsequently forms pores on the cell membrane, which causes the release of IL-1β and results in melanocyte pyroptosis. In our study, we utilize KPV-Lipos with <i>Nlrp3</i> shRNA to precisely knockdown NLRP3 expression in melanocytes and effectively alleviate vitiligo development, which provide a potentially promising strategy for the treatment of vitiligo. MC, melanocytes.</p></figure>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"74 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2025-09-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145032172","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
Proteasome 20S beta 8 (PSMB8) as a metabolic switcher of neuronal ferroptosis in multiple sclerosis 蛋白酶体20S β 8 (PSMB8)作为多发性硬化症中神经元铁质凋亡的代谢开关
IF 12.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-10 DOI: 10.1038/s41418-025-01572-x
Wei-Na Jin, Fu-Dong Shi
{"title":"Proteasome 20S beta 8 (PSMB8) as a metabolic switcher of neuronal ferroptosis in multiple sclerosis","authors":"Wei-Na Jin, Fu-Dong Shi","doi":"10.1038/s41418-025-01572-x","DOIUrl":"https://doi.org/10.1038/s41418-025-01572-x","url":null,"abstract":"","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"164 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145025489","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
RNF128 regulates the adaptive metabolic response to fasting by modulating PPARα function RNF128通过调节PPARα功能调节对禁食的适应性代谢反应
IF 12.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-10 DOI: 10.1038/s41418-025-01579-4
Yu-Lung Lin, Pei-Yao Liu, Yu-Ling Tsai, Chien-Ming Lin, Yu-Guang Chen, Jun-Ren Sun, Yu-Chan Chang, Wen-Chiuan Tsai, Yi-Xuan Ding, Chi-Wei Liu, Shih-Yun Wang, Ying-Chuan Chen

Peroxisome proliferator-activated receptor alpha (PPARα) is a crucial transcriptional factor that regulates fatty acid β-oxidation and ketogenesis in response to fasting. However, the mechanisms underlying PPARα function remain unclear. This study identified a novel PPARα-binding protein—RING finger protein 128 (RNF128)—that facilitates PPARα polyubiquitination, resulting in the degradation and suppression of PPARα function during fasting. Furthermore, RNF128 overexpression inhibited fibroblast growth factor 21 expression and lipid metabolism-related genes by facilitating PPARα degradation during fasting. In contrast, silencing RNF128 expression enhanced PPARα-dependent fatty acid β-oxidation and ketogenesis in starved cells. In vivo experiments demonstrated that RNF128 deficiency also significantly reduced lipid levels while increasing fatty acid β-oxidation and ketogenesis during fasting. Adeno-associated virus serotype 8-mediated RNF128 overexpression resulted in increased lipid levels and decreased expression of genes related to fatty acid β-oxidation and ketogenesis in fasted mice. Our findings revealed that RNF128 is crucial for metabolic adaptation to fasting in the liver by interacting with PPARα, thereby enhancing its polyubiquitination and degradation. Therefore, RNF128 is a novel regulator of PPARα function under nutrient-deprived conditions.

过氧化物酶体增殖物激活受体α (PPARα)是一个重要的转录因子,在禁食反应中调节脂肪酸β氧化和生酮。然而,PPARα功能的机制尚不清楚。本研究发现了一种新的PPARα结合蛋白-环指蛋白128 (RNF128) -促进PPARα多泛素化,导致禁食期间PPARα功能的降解和抑制。此外,RNF128过表达通过促进禁食期间PPARα降解,抑制成纤维细胞生长因子21的表达和脂质代谢相关基因。相反,沉默RNF128表达可增强饥饿细胞中ppar α-依赖性脂肪酸β-氧化和酮生成。体内实验表明,RNF128缺乏也显著降低了脂质水平,同时增加了禁食期间脂肪酸β氧化和生酮。腺相关病毒血清型8介导的RNF128过表达导致禁食小鼠脂质水平升高,脂肪酸β-氧化和生酮相关基因表达降低。我们的研究结果表明,RNF128通过与PPARα相互作用,从而增强其多泛素化和降解,对肝脏对禁食的代谢适应至关重要。因此,RNF128是营养剥夺条件下PPARα功能的新调节因子。
{"title":"RNF128 regulates the adaptive metabolic response to fasting by modulating PPARα function","authors":"Yu-Lung Lin, Pei-Yao Liu, Yu-Ling Tsai, Chien-Ming Lin, Yu-Guang Chen, Jun-Ren Sun, Yu-Chan Chang, Wen-Chiuan Tsai, Yi-Xuan Ding, Chi-Wei Liu, Shih-Yun Wang, Ying-Chuan Chen","doi":"10.1038/s41418-025-01579-4","DOIUrl":"https://doi.org/10.1038/s41418-025-01579-4","url":null,"abstract":"<p>Peroxisome proliferator-activated receptor alpha (PPARα) is a crucial transcriptional factor that regulates fatty acid β-oxidation and ketogenesis in response to fasting. However, the mechanisms underlying PPARα function remain unclear. This study identified a novel PPARα-binding protein—RING finger protein 128 (RNF128)—that facilitates PPARα polyubiquitination, resulting in the degradation and suppression of PPARα function during fasting. Furthermore, RNF128 overexpression inhibited fibroblast growth factor 21 expression and lipid metabolism-related genes by facilitating PPARα degradation during fasting. In contrast, silencing RNF128 expression enhanced PPARα-dependent fatty acid β-oxidation and ketogenesis in starved cells. In vivo experiments demonstrated that RNF128 deficiency also significantly reduced lipid levels while increasing fatty acid β-oxidation and ketogenesis during fasting. Adeno-associated virus serotype 8-mediated RNF128 overexpression resulted in increased lipid levels and decreased expression of genes related to fatty acid β-oxidation and ketogenesis in fasted mice. Our findings revealed that RNF128 is crucial for metabolic adaptation to fasting in the liver by interacting with PPARα, thereby enhancing its polyubiquitination and degradation. Therefore, RNF128 is a novel regulator of PPARα function under nutrient-deprived conditions.</p><figure></figure>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"24 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2025-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145025490","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
Cell death in multiple sclerosis 多发性硬化的细胞死亡
IF 12.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-09 DOI: 10.1038/s41418-025-01576-7
Shuzhen Guan, Huimin Zhu, Mengting Zhang, Fu-Dong Shi, Bo Yan

Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system (CNS) characterized by inflammatory demyelination and progressive neurodegeneration. Although current disease-modifying therapies modulate peripheral autoimmune responses, they are insufficient to fully prevent tissue specific neuroinflammation and long-term neuronal and oligodendrocyte loss. Growing evidence implicates various regulated cell death (RCD) pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis, not only as downstream consequences of chronic inflammation, but also as active drivers of demyelination, axonal injury, and glial dysfunction in MS. These RCD modalities contribute to MS pathology by disrupting cellular homeostasis and sustaining immune activation through the continuous release of damage-associated molecular patterns (DAMPs), thereby establishing a self-amplifying loop between cell death and inflammation. Furthermore, distinct RCD forms can co-occur within lesions, contributing to the complex cellular landscape of MS. This review summarizes current understanding of RCD mechanisms in MS, focusing on their contributions to neuroinflammation and neurodegeneration across different disease stages. We also discuss recent therapeutic advances targeting RCD, including approved drugs whose efficacy may be partially attributed to modulation of cell death, and emerging small-molecule inhibitors targeting key cell death components such as receptor-interacting protein kinase 1 (RIPK1) and NOD-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3). Targeting RCD in conjunction with inflammation may represent a more pragmatic approach for mitigating MS progression and neurodegeneration.

多发性硬化症(MS)是中枢神经系统(CNS)的一种慢性自身免疫性疾病,以炎症性脱髓鞘和进行性神经变性为特征。虽然目前的疾病修饰疗法可以调节外周自身免疫反应,但它们不足以完全预防组织特异性神经炎症和长期的神经元和少突胶质细胞损失。越来越多的证据表明,多种受调节的细胞死亡(RCD)途径,包括细胞凋亡、坏死坏死、焦亡和铁亡,不仅是慢性炎症的下游后果,而且是MS脱髓鞘、轴突损伤和神经胶质功能障碍的积极驱动因素。这些RCD方式通过破坏细胞稳态和持续释放损伤相关分子模式(DAMPs)来维持免疫激活,从而促进MS病理。从而在细胞死亡和炎症之间建立一个自我放大的循环。此外,不同形式的RCD可以在病变内同时发生,从而导致MS复杂的细胞景观。本文综述了目前对MS中RCD机制的理解,重点讨论了它们在不同疾病阶段对神经炎症和神经退行性变的影响。我们还讨论了最近针对RCD的治疗进展,包括已批准的药物,其疗效可能部分取决于细胞死亡的调节,以及针对关键细胞死亡成分的新出现的小分子抑制剂,如受体相互作用蛋白激酶1 (RIPK1)和NOD-、富含亮氨酸的重复序列-和含pyrin结构域的蛋白3 (NLRP3)。结合炎症靶向RCD可能是缓解MS进展和神经退行性变的更实用的方法。
{"title":"Cell death in multiple sclerosis","authors":"Shuzhen Guan, Huimin Zhu, Mengting Zhang, Fu-Dong Shi, Bo Yan","doi":"10.1038/s41418-025-01576-7","DOIUrl":"https://doi.org/10.1038/s41418-025-01576-7","url":null,"abstract":"<p>Multiple sclerosis (MS) is a chronic autoimmune disorder of the central nervous system (CNS) characterized by inflammatory demyelination and progressive neurodegeneration. Although current disease-modifying therapies modulate peripheral autoimmune responses, they are insufficient to fully prevent tissue specific neuroinflammation and long-term neuronal and oligodendrocyte loss. Growing evidence implicates various regulated cell death (RCD) pathways, including apoptosis, necroptosis, pyroptosis, and ferroptosis, not only as downstream consequences of chronic inflammation, but also as active drivers of demyelination, axonal injury, and glial dysfunction in MS. These RCD modalities contribute to MS pathology by disrupting cellular homeostasis and sustaining immune activation through the continuous release of damage-associated molecular patterns (DAMPs), thereby establishing a self-amplifying loop between cell death and inflammation. Furthermore, distinct RCD forms can co-occur within lesions, contributing to the complex cellular landscape of MS. This review summarizes current understanding of RCD mechanisms in MS, focusing on their contributions to neuroinflammation and neurodegeneration across different disease stages. We also discuss recent therapeutic advances targeting RCD, including approved drugs whose efficacy may be partially attributed to modulation of cell death, and emerging small-molecule inhibitors targeting key cell death components such as receptor-interacting protein kinase 1 (RIPK1) and NOD-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3). Targeting RCD in conjunction with inflammation may represent a more pragmatic approach for mitigating MS progression and neurodegeneration.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"39 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2025-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145017494","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
PRMT5 encourages cell migration and metastasis of tongue squamous cell carcinoma through methylating ΔNp63α PRMT5通过甲基化ΔNp63α促进舌鳞状细胞癌的细胞迁移和转移
IF 12.4 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-09-06 DOI: 10.1038/s41418-025-01575-8
Shijie Fan, Wen-juan Li, Yucheng Qi, Zejiao Li, Yao-hui He, Xiushuang Luo, Xiaoyun Nie, Jia Wang, Jinqi Ji, Haoran Tian, Yang Cao, Ya Hou, Ning Ji, Zhi-xiong Jim Xiao, Xiaobo Wang, Wen Liu, Chenghua Li

Tongue squamous cell carcinoma (TSCC) is a common oral malignancy prone to metastasis, whose underlying mechanism remains obscure. Here, we report the oncogenic roles of protein arginine methyltransferase 5 (PRMT5) in TSCC via inhibiting transcription factor ΔNp63α. We found that PRMT5 physically interacts with ΔNp63α, resulting in impairment of ΔNp63α-mediated transcriptional regulation. Further investigation revealed that PRMT5 is significantly upregulated in late stages of TSCC and correlated to poor prognosis. On the other hand, inhibition on ΔNp63α contributes to PRMT5-induced migration and metastasis of TSCC cells. Mechanistically, PRMT5 mediates methylation of ΔNp63α at Arg561, which facilitates CDK1-mediated phosphorylation of ΔNp63α and results in weakened DNA binding of this transcription factor. Consequently, ΔNp63α-mediated suppression on cell migration is attenuated in TSCC. Inhibition of PRMT5 efficiently restrain metastasis of TSCC cells in vivo. Our study is helpful to illuminate the molecular mechanism of TSCC metastasis and to provide a new therapeutic strategy for this malignancy.

舌鳞状细胞癌(TSCC)是一种常见的易转移的口腔恶性肿瘤,其发病机制尚不清楚。在这里,我们报告了蛋白精氨酸甲基转移酶5 (PRMT5)通过抑制转录因子ΔNp63α在TSCC中的致癌作用。我们发现PRMT5与ΔNp63α发生物理相互作用,导致ΔNp63α-mediated转录调控受损。进一步研究发现,PRMT5在TSCC晚期显著上调,并与预后不良相关。另一方面,抑制ΔNp63α有助于prmt5诱导的TSCC细胞迁移和转移。在机制上,PRMT5介导Arg561位点ΔNp63α的甲基化,从而促进cdk1介导的ΔNp63α磷酸化,并导致该转录因子的DNA结合减弱。因此,ΔNp63α-mediated对TSCC中细胞迁移的抑制减弱。体内抑制PRMT5可有效抑制TSCC细胞的转移。我们的研究有助于阐明TSCC转移的分子机制,并为这种恶性肿瘤的治疗提供新的策略。
{"title":"PRMT5 encourages cell migration and metastasis of tongue squamous cell carcinoma through methylating ΔNp63α","authors":"Shijie Fan, Wen-juan Li, Yucheng Qi, Zejiao Li, Yao-hui He, Xiushuang Luo, Xiaoyun Nie, Jia Wang, Jinqi Ji, Haoran Tian, Yang Cao, Ya Hou, Ning Ji, Zhi-xiong Jim Xiao, Xiaobo Wang, Wen Liu, Chenghua Li","doi":"10.1038/s41418-025-01575-8","DOIUrl":"https://doi.org/10.1038/s41418-025-01575-8","url":null,"abstract":"<p>Tongue squamous cell carcinoma (TSCC) is a common oral malignancy prone to metastasis, whose underlying mechanism remains obscure. Here, we report the oncogenic roles of protein arginine methyltransferase 5 (PRMT5) in TSCC via inhibiting transcription factor ΔNp63α. We found that PRMT5 physically interacts with ΔNp63α, resulting in impairment of ΔNp63α-mediated transcriptional regulation. Further investigation revealed that PRMT5 is significantly upregulated in late stages of TSCC and correlated to poor prognosis. On the other hand, inhibition on ΔNp63α contributes to PRMT5-induced migration and metastasis of TSCC cells. Mechanistically, PRMT5 mediates methylation of ΔNp63α at Arg561, which facilitates CDK1-mediated phosphorylation of ΔNp63α and results in weakened DNA binding of this transcription factor. Consequently, ΔNp63α-mediated suppression on cell migration is attenuated in TSCC. Inhibition of PRMT5 efficiently restrain metastasis of TSCC cells in vivo. Our study is helpful to illuminate the molecular mechanism of TSCC metastasis and to provide a new therapeutic strategy for this malignancy.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"14 1","pages":""},"PeriodicalIF":12.4,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145002907","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