首页 > 最新文献

Cell Death & Disease最新文献

英文 中文
Dual Bcl-2/Bcl-xl inhibition via AZD0466 combines with immune checkpoint blockade to enhance anti-tumour activity. 通过AZD0466双重抑制Bcl-2/Bcl-xl联合免疫检查点阻断增强抗肿瘤活性。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2026-01-06 DOI: 10.1038/s41419-025-08354-w
Dane M Newman, Courtney L Andersen, Leonie A Cluse, Andrea Newbold, Peter Fraser, Benjamin W C Legg, Luyao Kevin Xu, Deanna A Mele, Ricky W Johnstone

Small molecule inhibitors designed to specifically target oncogenic proteins have demonstrated potent anti-tumour activities due to direct effects on tumour cells survival and/or proliferation. However, the effects of these compounds on normal cells, specifically immune cells and their potential to impede or enhance anti-cancer immunotherapies has yet to be fully explored. Using an in vitro co-culture system to assess CD8+ T cell killing of tumour cells, we identified compounds that inhibit Bcl-2 and Bcl-xl as agents that can induce tumour cell death without impacting the differentiation or function of anti-tumour T cells. Accordingly, in vivo treatment of mice bearing solid tumours with a combination of the Bcl-2/Bcl-xl inhibitor AZD0466 and anti-PD-L1 immunotherapy resulted in enhanced anti-tumour effects and improved survival compared to equivalent monotherapies.

专为致癌蛋白设计的小分子抑制剂由于对肿瘤细胞存活和/或增殖的直接影响,已经证明了有效的抗肿瘤活性。然而,这些化合物对正常细胞,特别是免疫细胞的影响及其阻碍或增强抗癌免疫治疗的潜力尚未得到充分探索。使用体外共培养系统来评估CD8+ T细胞对肿瘤细胞的杀伤作用,我们发现抑制Bcl-2和Bcl-xl的化合物可以诱导肿瘤细胞死亡,而不影响抗肿瘤T细胞的分化或功能。因此,Bcl-2/Bcl-xl抑制剂AZD0466和抗pd - l1免疫疗法联合治疗实体瘤小鼠的体内治疗与等效的单一疗法相比,可增强抗肿瘤作用并提高生存率。
{"title":"Dual Bcl-2/Bcl-xl inhibition via AZD0466 combines with immune checkpoint blockade to enhance anti-tumour activity.","authors":"Dane M Newman, Courtney L Andersen, Leonie A Cluse, Andrea Newbold, Peter Fraser, Benjamin W C Legg, Luyao Kevin Xu, Deanna A Mele, Ricky W Johnstone","doi":"10.1038/s41419-025-08354-w","DOIUrl":"https://doi.org/10.1038/s41419-025-08354-w","url":null,"abstract":"<p><p>Small molecule inhibitors designed to specifically target oncogenic proteins have demonstrated potent anti-tumour activities due to direct effects on tumour cells survival and/or proliferation. However, the effects of these compounds on normal cells, specifically immune cells and their potential to impede or enhance anti-cancer immunotherapies has yet to be fully explored. Using an in vitro co-culture system to assess CD8+ T cell killing of tumour cells, we identified compounds that inhibit Bcl-2 and Bcl-xl as agents that can induce tumour cell death without impacting the differentiation or function of anti-tumour T cells. Accordingly, in vivo treatment of mice bearing solid tumours with a combination of the Bcl-2/Bcl-xl inhibitor AZD0466 and anti-PD-L1 immunotherapy resulted in enhanced anti-tumour effects and improved survival compared to equivalent monotherapies.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2026-01-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145910379","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
EIF4B Ser93 phosphorylation by ERK2 promotes epithelial-mesenchymal transition to drive colorectal cancer metastasis. ERK2磷酸化EIF4B Ser93促进上皮-间质转化,驱动结直肠癌转移。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2026-01-05 DOI: 10.1038/s41419-025-08375-5
Siqi Wen, Min Lin, Man Zhang, Zhao Li, Jinchi Chen, Bei Yi, Dejun Liu, Ruiqi Chen, Tianyu Chen, Rong Liang, Wei Jiang

Colorectal cancer (CRC) is the third most common malignant tumor and the second leading cause of cancer-related mortality globally. Epithelial to mesenchymal transition (EMT) contributes to CRC metastasis and poor prognosis. Aberrant protein phosphorylation is implicated in CRC progression, warranting further investigation into its molecular mechanisms. Herein, we have identified significant alterations in protein phosphorylation associated with CRC through tandem mass tag (TMT) label-based phosphoproteomic analysis. The functions and enriched signaling pathways of these proteins were predominantly linked to the EMT process. Notably, the phosphorylation of eIF4B at Ser93 exhibited the most pronounced increase in CRC, a finding that was further validated in CRC tissues and cell lines by a newly generated antibody targeting eIF4B Ser93 phosphorylation. Phosphorylation of eIF4B Ser93 promoted CRC progression and metastasis both in vitro and in vivo. Mechanistically, eIF4B Ser93 phosphorylation decreased ubiquitination-mediated eIF4B degradation and enhanced its translation activity, through which it facilitated the translation of mesenchymal markers. Additionally, ERK2 directly phosphorylated eIF4B at Ser93, while inhibiting this phosphorylation is essential for the anti-cancer efficacy of the ERK2 inhibitor, Vx-11e. Together, the phosphorylation of eIF4B Ser93 driven by ERK2 promotes CRC growth and metastasis through the activation of EMT. Our findings indicate a novel therapeutic target and provide promising strategies for clinical intervention in human CRC.

结直肠癌(CRC)是全球第三大最常见的恶性肿瘤,也是导致癌症相关死亡的第二大原因。上皮细胞向间质转化(EMT)有助于结直肠癌的转移和不良预后。异常蛋白磷酸化与结直肠癌的进展有关,需要进一步研究其分子机制。在此,我们通过串联质量标签(TMT)标记为基础的磷酸化蛋白质组学分析发现了与CRC相关的蛋白磷酸化的显著变化。这些蛋白的功能和丰富的信号通路主要与EMT过程有关。值得注意的是,eIF4B Ser93位点的磷酸化在结直肠癌中表现出最明显的增加,这一发现在结直肠癌组织和细胞系中被一种新产生的靶向eIF4B Ser93磷酸化的抗体进一步证实。在体外和体内,eIF4B Ser93的磷酸化促进了结直肠癌的进展和转移。从机制上讲,eIF4B Ser93磷酸化降低了泛素化介导的eIF4B降解,增强了其翻译活性,从而促进了间质标记物的翻译。此外,ERK2直接使eIF4B在Ser93位点磷酸化,而抑制这种磷酸化对于ERK2抑制剂Vx-11e的抗癌功效至关重要。总之,ERK2驱动的eIF4B Ser93磷酸化通过激活EMT促进结直肠癌的生长和转移。我们的发现为人类结直肠癌的临床干预提供了新的治疗靶点和有希望的策略。
{"title":"EIF4B Ser93 phosphorylation by ERK2 promotes epithelial-mesenchymal transition to drive colorectal cancer metastasis.","authors":"Siqi Wen, Min Lin, Man Zhang, Zhao Li, Jinchi Chen, Bei Yi, Dejun Liu, Ruiqi Chen, Tianyu Chen, Rong Liang, Wei Jiang","doi":"10.1038/s41419-025-08375-5","DOIUrl":"https://doi.org/10.1038/s41419-025-08375-5","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is the third most common malignant tumor and the second leading cause of cancer-related mortality globally. Epithelial to mesenchymal transition (EMT) contributes to CRC metastasis and poor prognosis. Aberrant protein phosphorylation is implicated in CRC progression, warranting further investigation into its molecular mechanisms. Herein, we have identified significant alterations in protein phosphorylation associated with CRC through tandem mass tag (TMT) label-based phosphoproteomic analysis. The functions and enriched signaling pathways of these proteins were predominantly linked to the EMT process. Notably, the phosphorylation of eIF4B at Ser93 exhibited the most pronounced increase in CRC, a finding that was further validated in CRC tissues and cell lines by a newly generated antibody targeting eIF4B Ser93 phosphorylation. Phosphorylation of eIF4B Ser93 promoted CRC progression and metastasis both in vitro and in vivo. Mechanistically, eIF4B Ser93 phosphorylation decreased ubiquitination-mediated eIF4B degradation and enhanced its translation activity, through which it facilitated the translation of mesenchymal markers. Additionally, ERK2 directly phosphorylated eIF4B at Ser93, while inhibiting this phosphorylation is essential for the anti-cancer efficacy of the ERK2 inhibitor, Vx-11e. Together, the phosphorylation of eIF4B Ser93 driven by ERK2 promotes CRC growth and metastasis through the activation of EMT. Our findings indicate a novel therapeutic target and provide promising strategies for clinical intervention in human CRC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2026-01-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145905718","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
Damage-induced pyroptosis drives endogenous thymic regeneration by activating the purinergic receptor P2Y2. 损伤诱导的焦亡通过激活嘌呤能受体P2Y2驱动内源性胸腺再生。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2026-01-03 DOI: 10.1038/s41419-025-08345-x
Sinéad Kinsella, Cindy A Evandy, Kirsten Cooper, Erin Kirsche, Makya Warren, Paul deRoos, Antonella Cardinale, Lorenzo Iovino, David Granadier, Colton W Smith, Kayla Hopwo, Lucas B Sullivan, Enrico Velardi, Jarrod A Dudakov

T cell recovery is critical following damage, such as hematopoietic cell transplantation (HCT), with increased reconstitution associated with improved clinical outcomes. Endogenous thymic regeneration, a crucial process for restoring immune competence following cytoreductive therapies such as HCT conditioning, is often delayed, limiting T cell reconstitution. Fully understanding the molecular mechanisms driving regeneration is therefore crucial for uncovering therapeutic targets that can be exploited to enhance thymic function. Here, we identified that CD4+ CD8+ thymocytes rapidly and acutely undergo lytic cell death, specifically pyroptosis, following acute damage caused by ionizing radiation, and release damage-associated molecular patterns (DAMPS) into the thymic microenvironment, including ATP. Extracellular ATP stimulates the P2Y2 purinergic receptor on thymic epithelial cells (TECs)-a stromal cell crucial for supporting T cell development-resulting in the upregulation FOXN1, the master TEC transcription factor. Targeting the P2Y2 receptor with a P2Y2 agonist, UTPγS, promotes rapid regeneration of the TEC compartment in vivo following acute damage. These findings reveal a novel damage-sensing mechanism employed by the thymus where thymocytes adopt an alternative cell death mechanism which promotes thymic repair via P2Y2 signaling in TECs. This work identifies P2Y2 as a promising therapeutic target for enhancing thymus regeneration and improving immune recovery after HCT.

T细胞恢复是损伤后的关键,如造血细胞移植(HCT),增加重建与改善临床结果相关。内源性胸腺再生是细胞减少疗法(如HCT调节)后恢复免疫能力的关键过程,但往往延迟,限制了T细胞的重建。因此,充分了解驱动再生的分子机制对于发现可用于增强胸腺功能的治疗靶点至关重要。在这里,我们发现CD4+ CD8+胸腺细胞在电离辐射引起的急性损伤后迅速而急性地发生溶解性细胞死亡,特别是焦亡,并将损伤相关分子模式(DAMPS)释放到胸腺微环境中,包括ATP。细胞外ATP刺激胸腺上皮细胞(TECs)上的P2Y2嘌呤能受体(一种对支持T细胞发育至关重要的基质细胞),导致TEC主转录因子FOXN1上调。用P2Y2受体激动剂UTPγS靶向P2Y2受体,可促进体内TEC室在急性损伤后的快速再生。这些发现揭示了胸腺采用的一种新的损伤感知机制,其中胸腺细胞采用另一种细胞死亡机制,通过tec中的P2Y2信号传导促进胸腺修复。这项工作确定P2Y2是一个有希望的治疗靶点,可以增强HCT后胸腺再生和改善免疫恢复。
{"title":"Damage-induced pyroptosis drives endogenous thymic regeneration by activating the purinergic receptor P2Y2.","authors":"Sinéad Kinsella, Cindy A Evandy, Kirsten Cooper, Erin Kirsche, Makya Warren, Paul deRoos, Antonella Cardinale, Lorenzo Iovino, David Granadier, Colton W Smith, Kayla Hopwo, Lucas B Sullivan, Enrico Velardi, Jarrod A Dudakov","doi":"10.1038/s41419-025-08345-x","DOIUrl":"10.1038/s41419-025-08345-x","url":null,"abstract":"<p><p>T cell recovery is critical following damage, such as hematopoietic cell transplantation (HCT), with increased reconstitution associated with improved clinical outcomes. Endogenous thymic regeneration, a crucial process for restoring immune competence following cytoreductive therapies such as HCT conditioning, is often delayed, limiting T cell reconstitution. Fully understanding the molecular mechanisms driving regeneration is therefore crucial for uncovering therapeutic targets that can be exploited to enhance thymic function. Here, we identified that CD4+ CD8+ thymocytes rapidly and acutely undergo lytic cell death, specifically pyroptosis, following acute damage caused by ionizing radiation, and release damage-associated molecular patterns (DAMPS) into the thymic microenvironment, including ATP. Extracellular ATP stimulates the P2Y2 purinergic receptor on thymic epithelial cells (TECs)-a stromal cell crucial for supporting T cell development-resulting in the upregulation FOXN1, the master TEC transcription factor. Targeting the P2Y2 receptor with a P2Y2 agonist, UTPγS, promotes rapid regeneration of the TEC compartment in vivo following acute damage. These findings reveal a novel damage-sensing mechanism employed by the thymus where thymocytes adopt an alternative cell death mechanism which promotes thymic repair via P2Y2 signaling in TECs. This work identifies P2Y2 as a promising therapeutic target for enhancing thymus regeneration and improving immune recovery after HCT.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896360","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
SOX4-STAT6-MTHFD2 axis drives hepatocellular carcinoma progression and treatment resistance. SOX4-STAT6-MTHFD2轴驱动肝细胞癌进展和治疗耐药性。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2026-01-03 DOI: 10.1038/s41419-025-08394-2
Chia-Lung Tsai, Ming-Chin Yu, Cheng-Lung Hsu, Hsiang-Yu Tang, Yun-Shien Lee, Lang-Ming Chi, Sey-En Lin, Mei-Ling Cheng, Heng-Yuan Hsu, Chi-Neu Tsai

Hepatocellular carcinoma (HCC) is a major global health burden. Despite recent advances in immunotherapy, tyrosine kinase inhibitors (TKIs) treatment or combined therapies, therapeutic resistance and disease progression remain significant challenges. SOX4, a transcription factor frequently overexpressed in HCC and other cancers, has been linked to drug resistance and poor prognosis; however, the underlying molecular mechanisms remain unexplored. In this study, we identify STAT6 as a novel transcriptional target and interacting partner of SOX4 in HCC cells. Genetic ablation or knockdown of SOX4 induced hypermethylation of the STAT6 promoter, suppressing its expression, while treatment with the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine restored STAT6 levels, indicating an epigenetic mechanism of regulation. In addition, SOX4 is physically associated with STAT6, as confirmed by co-immunoprecipitation and immunofluorescence. SOX4 depletion impaired interleukin-4 (IL-4)-induced phosphorylation of STAT6 at tyrosine residue 641 (Y641), implicating SOX4 in IL-4-mediated STAT6 activation. Chromatin immunoprecipitation (ChIP) assays demonstrated that SOX4 and STAT6 co-occupy the promoter of MTHFD2, a key enzyme in folate metabolism, regulating NADH/NADPH production and nucleotide biosynthesis. Knockdown of SOX4 or STAT6, or mutation of their binding sites within the MTHFD2 promoter, reduced MTHFD2 expression, NADPH levels, and nucleotide synthesis. Transcriptomic analyses from TCGA-LIHC and our independent cohort revealed a strong positive correlation between SOX4, STAT6, and MTHFD2, with MTHFD2 overexpression linked to poor overall survival. Clinically, elevated SOX4/STAT6/MTHFD2 axis activity was associated with resistance to immunotherapy or TKIs, either in our enrolled cohort or transcriptome data obtained from GSE109211. Metabolomic profiling further revealed increased NADPH and nucleotide biosynthesis in tumors with high SOX4/STAT6/MTHFD2 expression. Targeting STAT6 or MTHFD2 suppressed tumor growth in TKIs-resistant patient-derived xenograft models. Collectively, our findings identify the SOX4-STAT6-MTHFD2 axis as a critical driver of HCC progression and therapeutic resistance, offering a promising target for intervention in refractory HCC.

肝细胞癌(HCC)是全球主要的健康负担。尽管最近在免疫治疗、酪氨酸激酶抑制剂(TKIs)治疗或联合治疗方面取得了进展,但治疗耐药性和疾病进展仍然是重大挑战。SOX4是一种在HCC和其他癌症中经常过度表达的转录因子,与耐药和预后不良有关;然而,潜在的分子机制仍未被探索。在本研究中,我们发现STAT6是HCC细胞中SOX4的一个新的转录靶点和相互作用伙伴。基因切除或敲低SOX4诱导STAT6启动子的超甲基化,抑制其表达,而DNA甲基转移酶抑制剂5-Aza-2'-脱氧胞苷治疗恢复STAT6水平,表明调控的表观遗传机制。此外,通过免疫共沉淀和免疫荧光证实,SOX4与STAT6存在物理关联。SOX4缺失破坏了白细胞介素-4 (IL-4)诱导的酪氨酸残基641 (Y641)处STAT6的磷酸化,暗示SOX4参与了IL-4介导的STAT6激活。染色质免疫沉淀(ChIP)实验表明,SOX4和STAT6共同占据MTHFD2启动子,MTHFD2是叶酸代谢的关键酶,调节NADH/NADPH的产生和核苷酸的生物合成。敲低SOX4或STAT6,或MTHFD2启动子内它们的结合位点发生突变,可降低MTHFD2表达、NADPH水平和核苷酸合成。来自TCGA-LIHC和我们独立队列的转录组学分析显示,SOX4、STAT6和MTHFD2之间存在很强的正相关,MTHFD2过表达与较差的总生存率有关。在临床上,SOX4/STAT6/MTHFD2轴活性升高与免疫治疗或TKIs耐药相关,无论是在我们的入组队列中还是从GSE109211获得的转录组数据中都是如此。代谢组学分析进一步显示,在SOX4/STAT6/MTHFD2高表达的肿瘤中,NADPH和核苷酸生物合成增加。靶向STAT6或MTHFD2抑制tkis耐药患者来源的异种移植模型的肿瘤生长。总之,我们的研究结果确定SOX4-STAT6-MTHFD2轴是HCC进展和治疗耐药的关键驱动因素,为难治性HCC的干预提供了一个有希望的靶点。
{"title":"SOX4-STAT6-MTHFD2 axis drives hepatocellular carcinoma progression and treatment resistance.","authors":"Chia-Lung Tsai, Ming-Chin Yu, Cheng-Lung Hsu, Hsiang-Yu Tang, Yun-Shien Lee, Lang-Ming Chi, Sey-En Lin, Mei-Ling Cheng, Heng-Yuan Hsu, Chi-Neu Tsai","doi":"10.1038/s41419-025-08394-2","DOIUrl":"https://doi.org/10.1038/s41419-025-08394-2","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) is a major global health burden. Despite recent advances in immunotherapy, tyrosine kinase inhibitors (TKIs) treatment or combined therapies, therapeutic resistance and disease progression remain significant challenges. SOX4, a transcription factor frequently overexpressed in HCC and other cancers, has been linked to drug resistance and poor prognosis; however, the underlying molecular mechanisms remain unexplored. In this study, we identify STAT6 as a novel transcriptional target and interacting partner of SOX4 in HCC cells. Genetic ablation or knockdown of SOX4 induced hypermethylation of the STAT6 promoter, suppressing its expression, while treatment with the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine restored STAT6 levels, indicating an epigenetic mechanism of regulation. In addition, SOX4 is physically associated with STAT6, as confirmed by co-immunoprecipitation and immunofluorescence. SOX4 depletion impaired interleukin-4 (IL-4)-induced phosphorylation of STAT6 at tyrosine residue 641 (Y641), implicating SOX4 in IL-4-mediated STAT6 activation. Chromatin immunoprecipitation (ChIP) assays demonstrated that SOX4 and STAT6 co-occupy the promoter of MTHFD2, a key enzyme in folate metabolism, regulating NADH/NADPH production and nucleotide biosynthesis. Knockdown of SOX4 or STAT6, or mutation of their binding sites within the MTHFD2 promoter, reduced MTHFD2 expression, NADPH levels, and nucleotide synthesis. Transcriptomic analyses from TCGA-LIHC and our independent cohort revealed a strong positive correlation between SOX4, STAT6, and MTHFD2, with MTHFD2 overexpression linked to poor overall survival. Clinically, elevated SOX4/STAT6/MTHFD2 axis activity was associated with resistance to immunotherapy or TKIs, either in our enrolled cohort or transcriptome data obtained from GSE109211. Metabolomic profiling further revealed increased NADPH and nucleotide biosynthesis in tumors with high SOX4/STAT6/MTHFD2 expression. Targeting STAT6 or MTHFD2 suppressed tumor growth in TKIs-resistant patient-derived xenograft models. Collectively, our findings identify the SOX4-STAT6-MTHFD2 axis as a critical driver of HCC progression and therapeutic resistance, offering a promising target for intervention in refractory HCC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896323","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
MCJ modulates mitochondrial ETC flux to promote lipid metabolism-driven enhancement of cell proliferation and migration. MCJ调节线粒体ETC通量,促进脂质代谢驱动的细胞增殖和迁移增强。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2026-01-02 DOI: 10.1038/s41419-025-08398-y
Priyadarshika Pradhan, Tanvi Chaudhary, Shivali Mishra, Peter Konik, Eva Durinova, Roman Tuma, Abhijt De, Devanjan Sinha

Mitochondrial metabolism plays a crucial role in cancer progression and is associated with effective channeling of electrons through Complex I. The ability to adapt this electron flow as per cellular demands is critical for energy homeostasis. Our observations suggest that proliferating cells regulate the electron entry point through alterations in the levels of Methylation-Controlled J-protein (MCJ). Elevated MCJ levels were found to promote aggressive proliferative and migratory phenotypes, leading to increased primary tumor burden. The phenotype was attributed to MCJ-mediated regulation of mitochondrial bioenergetic plasticity, enabling a preferential rerouting of electron flux through succinate dehydrogenase complex (Complex II). Consequently, cells exhibited suppressed glycolysis and a metabolic shift toward lipid-fueled mitochondrial respiration, marked by increased lipid accumulation and its oxidation. Despite Complex I uncoupling, these cells maintained better respiratory output and preserved NADH levels to support an increased redox potential. These findings decouple the reliance on Complex I for effective mitochondrial respiration and underscore the significance of Complex II-driven metabolism in tumor growth, an important consideration for development of future therapeutics, particularly when current strategies predominantly target Complex I-dependent respiration.

线粒体代谢在癌症进展中起着至关重要的作用,并与电子通过复合体i的有效通道有关。根据细胞需求适应这种电子流的能力对能量稳态至关重要。我们的观察结果表明,增殖细胞通过改变甲基化控制j蛋白(MCJ)的水平来调节电子进入点。MCJ水平升高可促进侵袭性增殖和迁移表型,导致原发性肿瘤负担增加。该表型归因于mcj介导的线粒体生物能量可塑性调节,使电子通量通过琥珀酸脱氢酶复合物(复合物II)优先改变路线。因此,细胞表现出糖酵解抑制和向脂质驱动的线粒体呼吸代谢转变,其特征是脂质积累和氧化增加。尽管复合体I解偶联,这些细胞保持了更好的呼吸输出和NADH水平,以支持增加的氧化还原电位。这些发现解除了对复合体I有效线粒体呼吸的依赖,并强调了复合体ii驱动的代谢在肿瘤生长中的重要性,这是未来治疗方法发展的重要考虑因素,特别是当当前策略主要针对复合体I依赖的呼吸时。
{"title":"MCJ modulates mitochondrial ETC flux to promote lipid metabolism-driven enhancement of cell proliferation and migration.","authors":"Priyadarshika Pradhan, Tanvi Chaudhary, Shivali Mishra, Peter Konik, Eva Durinova, Roman Tuma, Abhijt De, Devanjan Sinha","doi":"10.1038/s41419-025-08398-y","DOIUrl":"https://doi.org/10.1038/s41419-025-08398-y","url":null,"abstract":"<p><p>Mitochondrial metabolism plays a crucial role in cancer progression and is associated with effective channeling of electrons through Complex I. The ability to adapt this electron flow as per cellular demands is critical for energy homeostasis. Our observations suggest that proliferating cells regulate the electron entry point through alterations in the levels of Methylation-Controlled J-protein (MCJ). Elevated MCJ levels were found to promote aggressive proliferative and migratory phenotypes, leading to increased primary tumor burden. The phenotype was attributed to MCJ-mediated regulation of mitochondrial bioenergetic plasticity, enabling a preferential rerouting of electron flux through succinate dehydrogenase complex (Complex II). Consequently, cells exhibited suppressed glycolysis and a metabolic shift toward lipid-fueled mitochondrial respiration, marked by increased lipid accumulation and its oxidation. Despite Complex I uncoupling, these cells maintained better respiratory output and preserved NADH levels to support an increased redox potential. These findings decouple the reliance on Complex I for effective mitochondrial respiration and underscore the significance of Complex II-driven metabolism in tumor growth, an important consideration for development of future therapeutics, particularly when current strategies predominantly target Complex I-dependent respiration.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896325","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
CYP8B1 inhibits hepatocellular carcinoma progression by repressing PAK4 transcription through inhibition of nuclear translocation of u-STAT1. CYP8B1通过抑制u-STAT1核易位抑制PAK4转录,从而抑制肝癌进展。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-31 DOI: 10.1038/s41419-025-08393-3
Le Xu, Lin Xiong, Jingping Yuan, Yukai Chen, Fangfang Chen, Siyu Wang, Ximing Xu

Primary and secondary bile acid (BA) levels are elevated in patients with hepatocellular carcinoma (HCC). BAs are important signaling molecules that regulate CYP8B1 expression by targeting nuclear and membrane receptors. In this study, we aimed to determine the function of CYP8B1 in HCC. Examination of HCC tissue and bioinformatic analysis revealed that CYP8B1 expression is downregulated in HCC tissues and is associated with good prognosis. Cholic acid promoted Huh7 cell proliferation and migration by inhibiting CYP8B1 expression. Both in vitro and in vivo, CYP8B1 inhibited the proliferation, invasion, and migration of HCC cells. Nanopore long-read RNA-sequencing analysis identified PAK4 as a potential target of CYP8B1, and the MAPK pathway was associated with CYP8B1 expression. CYP8B1 inhibited PAK4 expression and Raf/MEK/ERK phosphorylation. Tissue microarray analysis also verified a strong correlation between CYP8B1 and PAK4 expression. In vitro Cell Counting Kit 8 assays and in vivo orthotopic liver tumor model analyses showed that CYP8B1 restores sorafenib sensitivity in resistant HC, suggesting its potential as a therapeutic target. IP-MS of CYP8B1 and transcription factor prediction of PAK4 revealed STAT1 as a potential transcription factor for PAK4, which may directly bind to CYP8B1. Chromatin immunoprecipitation confirmed that u-STAT1 directly binds to the PAK4 promoter, not p-STAT1. Overall, CYP8B1 binds to u-STAT1 in the cytoplasm, reducing the translocation of u-STAT1 from the cytoplasm to the nucleus, thereby inhibiting the transcription of PAK4 and ultimately inhibiting the phosphorylation of Raf/MEK/ERK. Our findings indicate that the CYP8B1/PAK4 axis is important in HCC progression and elucidate the mechanism by which BAs promote HCC. Thus, CYP8B1 is a potential therapeutic target for the clinical treatment of HCC.

原发性和继发性胆汁酸(BA)水平在肝细胞癌(HCC)患者中升高。BAs是通过靶向核和膜受体调控CYP8B1表达的重要信号分子。在本研究中,我们旨在确定CYP8B1在HCC中的功能。HCC组织检查和生物信息学分析显示,CYP8B1在HCC组织中表达下调,与预后良好相关。胆酸通过抑制CYP8B1表达促进Huh7细胞增殖和迁移。在体外和体内实验中,CYP8B1均能抑制HCC细胞的增殖、侵袭和迁移。纳米孔长读rna测序分析发现PAK4是CYP8B1的潜在靶点,MAPK通路与CYP8B1表达相关。CYP8B1抑制PAK4表达和Raf/MEK/ERK磷酸化。组织芯片分析也证实了CYP8B1与PAK4表达之间的强相关性。体外细胞计数试剂盒8检测和体内原位肝肿瘤模型分析显示,CYP8B1可恢复耐药HC中索拉非尼的敏感性,表明其可能是一种治疗靶点。CYP8B1的IP-MS和PAK4的转录因子预测显示STAT1是PAK4的潜在转录因子,可能直接结合CYP8B1。染色质免疫沉淀证实u-STAT1直接结合PAK4启动子,而不是p-STAT1。总的来说,CYP8B1与细胞质中的u-STAT1结合,减少u-STAT1从细胞质向细胞核的易位,从而抑制PAK4的转录,最终抑制Raf/MEK/ERK的磷酸化。我们的研究结果表明CYP8B1/PAK4轴在HCC进展中很重要,并阐明了BAs促进HCC的机制。因此,CYP8B1是HCC临床治疗的潜在治疗靶点。
{"title":"CYP8B1 inhibits hepatocellular carcinoma progression by repressing PAK4 transcription through inhibition of nuclear translocation of u-STAT1.","authors":"Le Xu, Lin Xiong, Jingping Yuan, Yukai Chen, Fangfang Chen, Siyu Wang, Ximing Xu","doi":"10.1038/s41419-025-08393-3","DOIUrl":"https://doi.org/10.1038/s41419-025-08393-3","url":null,"abstract":"<p><p>Primary and secondary bile acid (BA) levels are elevated in patients with hepatocellular carcinoma (HCC). BAs are important signaling molecules that regulate CYP8B1 expression by targeting nuclear and membrane receptors. In this study, we aimed to determine the function of CYP8B1 in HCC. Examination of HCC tissue and bioinformatic analysis revealed that CYP8B1 expression is downregulated in HCC tissues and is associated with good prognosis. Cholic acid promoted Huh7 cell proliferation and migration by inhibiting CYP8B1 expression. Both in vitro and in vivo, CYP8B1 inhibited the proliferation, invasion, and migration of HCC cells. Nanopore long-read RNA-sequencing analysis identified PAK4 as a potential target of CYP8B1, and the MAPK pathway was associated with CYP8B1 expression. CYP8B1 inhibited PAK4 expression and Raf/MEK/ERK phosphorylation. Tissue microarray analysis also verified a strong correlation between CYP8B1 and PAK4 expression. In vitro Cell Counting Kit 8 assays and in vivo orthotopic liver tumor model analyses showed that CYP8B1 restores sorafenib sensitivity in resistant HC, suggesting its potential as a therapeutic target. IP-MS of CYP8B1 and transcription factor prediction of PAK4 revealed STAT1 as a potential transcription factor for PAK4, which may directly bind to CYP8B1. Chromatin immunoprecipitation confirmed that u-STAT1 directly binds to the PAK4 promoter, not p-STAT1. Overall, CYP8B1 binds to u-STAT1 in the cytoplasm, reducing the translocation of u-STAT1 from the cytoplasm to the nucleus, thereby inhibiting the transcription of PAK4 and ultimately inhibiting the phosphorylation of Raf/MEK/ERK. Our findings indicate that the CYP8B1/PAK4 axis is important in HCC progression and elucidate the mechanism by which BAs promote HCC. Thus, CYP8B1 is a potential therapeutic target for the clinical treatment of HCC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145877910","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
CRX is an intrinsic suppressor of epithelial‒mesenchymal transition in retinal pigment epithelial cells: a promising therapeutic avenue for subretinal fibrosis. CRX是视网膜色素上皮细胞上皮-间质转化的内在抑制因子:视网膜下纤维化的一种有前景的治疗途径。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-31 DOI: 10.1038/s41419-025-08352-y
Dongli Li, Qingjian Ou, Furong Gao, Xi Wang, Lilin Zhu, Ye Zhou, Jing-Ying Xu, Caixia Jin, Juan Wang, Jieping Zhang, Jiao Li, Yanlong Bi, Lixia Lu, Guo-Tong Xu, Haibin Tian

The epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is one of the significant pathogenic mechanisms for the formation of subretinal fibrosis in age-related macular degeneration (AMD). Multiple signaling pathways that promote EMT have been well described, yet the endogenous signaling pathways that inhibit EMT within RPE cells remain largely elusive. In this study, we confirmed the expression of CRX in human RPE cells and human embryonic stem cell-derived RPE (ESC-RPE) cells. By employing sub-culture to disrupt intercellular connections and thereby inhibit the Hippo signaling pathway, combined with TGF-β1 treatment in vitro to mimic the microenvironment for the formation of subretinal fibrosis, it was revealed that Hippo/YAP1 and TGF-β1 synergistically promoted the nuclear translocation of β-catenin, and the latter bound to TCF7 to inhibit the expression of CRX. Overexpression of CRX was capable of suppressing the occurrence of EMT in ESC-RPE cells. CRX exerted its inhibitory effect on EMT partly by upregulating the expression of PPP2R2B. In the laser-induced choroidal neovascularization mouse model, the nuclear translocation of CRX took place in RPE cells, and overexpression of CRX played an inhibitory role in the formation of subretinal fibrosis. This study has identified CRX as an endogenous signaling molecule that inhibits EMT in RPE cells and has provided a new research target and treatment strategy for the treatment of wet AMD and the inhibition of subretinal fibrosis formation.

视网膜色素上皮(RPE)细胞的上皮-间质转化(EMT)是老年性黄斑变性(AMD)视网膜下纤维化形成的重要致病机制之一。促进EMT的多种信号通路已被很好地描述,但抑制RPE细胞内EMT的内源性信号通路仍在很大程度上难以捉摸。在本研究中,我们证实了CRX在人RPE细胞和人胚胎干细胞衍生的RPE (ESC-RPE)细胞中的表达。通过传代培养破坏细胞间连接从而抑制Hippo信号通路,结合TGF-β1体外处理模拟视网膜下纤维化形成的微环境,发现Hippo/YAP1和TGF-β1协同促进β-catenin的核易位,后者结合TCF7抑制CRX的表达。过表达CRX能够抑制ESC-RPE细胞中EMT的发生。CRX对EMT的抑制作用部分是通过上调PPP2R2B的表达来实现的。在激光诱导脉络膜新生血管小鼠模型中,RPE细胞中发生了CRX的核易位,CRX的过表达对视网膜下纤维化的形成起抑制作用。本研究发现CRX是抑制RPE细胞EMT的内源性信号分子,为治疗湿性AMD和抑制视网膜下纤维化形成提供了新的研究靶点和治疗策略。
{"title":"CRX is an intrinsic suppressor of epithelial‒mesenchymal transition in retinal pigment epithelial cells: a promising therapeutic avenue for subretinal fibrosis.","authors":"Dongli Li, Qingjian Ou, Furong Gao, Xi Wang, Lilin Zhu, Ye Zhou, Jing-Ying Xu, Caixia Jin, Juan Wang, Jieping Zhang, Jiao Li, Yanlong Bi, Lixia Lu, Guo-Tong Xu, Haibin Tian","doi":"10.1038/s41419-025-08352-y","DOIUrl":"https://doi.org/10.1038/s41419-025-08352-y","url":null,"abstract":"<p><p>The epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is one of the significant pathogenic mechanisms for the formation of subretinal fibrosis in age-related macular degeneration (AMD). Multiple signaling pathways that promote EMT have been well described, yet the endogenous signaling pathways that inhibit EMT within RPE cells remain largely elusive. In this study, we confirmed the expression of CRX in human RPE cells and human embryonic stem cell-derived RPE (ESC-RPE) cells. By employing sub-culture to disrupt intercellular connections and thereby inhibit the Hippo signaling pathway, combined with TGF-β1 treatment in vitro to mimic the microenvironment for the formation of subretinal fibrosis, it was revealed that Hippo/YAP1 and TGF-β1 synergistically promoted the nuclear translocation of β-catenin, and the latter bound to TCF7 to inhibit the expression of CRX. Overexpression of CRX was capable of suppressing the occurrence of EMT in ESC-RPE cells. CRX exerted its inhibitory effect on EMT partly by upregulating the expression of PPP2R2B. In the laser-induced choroidal neovascularization mouse model, the nuclear translocation of CRX took place in RPE cells, and overexpression of CRX played an inhibitory role in the formation of subretinal fibrosis. This study has identified CRX as an endogenous signaling molecule that inhibits EMT in RPE cells and has provided a new research target and treatment strategy for the treatment of wet AMD and the inhibition of subretinal fibrosis formation.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145862316","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
GATOR1 complex controls cisplatin sensitivity. GATOR1复合物控制顺铂敏感性。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-30 DOI: 10.1038/s41419-025-08392-4
Zhenrui Pan, Hanxiao Zhang, Xia Xiao, Catherine Brenner, Svetlana Dokudovskaya

Cisplatin administration is the primary chemotherapy approach for many epithelial cancers. However, resistance to this drug poses a significant challenge to effective treatment. Despite the identification of numerous factors associated with resistance, reliable biomarkers predicting drug response remain elusive. Previously, low expression of the NPRL2 tumor suppressor was linked to cisplatin resistance. NPRL2, along with NPRL3 and DEPDC5, forms the GATOR1 complex, an upstream regulator of the mTORС1, the function of which is perturbed in many cancers, particularly those resistant to cisplatin. Here, we compare non-cancerous bronchial epithelium BEAS-2B cells with GATOR1 deletions, serving as a model of intrinsic cisplatin resistance, with non-small cell lung cancer lines A549, H460, and H1975 with acquired resistance to the drug. We found that deletion of any GATOR1 member, not solely NPRL2, promotes cisplatin resistance, whereas their overexpression renders cells sensitive to the drug. In cells with GATOR1 deletions, expression of the ATP7A transporter required for cisplatin efflux is increased, while expression of cisplatin influx transporters CTR2 and LRRC8A is downregulated, especially after treatment with the drug. This hinders drug accumulation in cells, resulting in the formation of fewer cisplatin-DNA adducts. Simultaneously, these cells exhibit enhanced DNA damage response and mTORC1 activity. Overexpression of GATOR1 components and/or concomitant treatment with an mTORC1 inhibitor restores sensitivity to cisplatin. Transcriptomic analysis of GATOR1-deleted BEAS-2B cells, treated or not with the drug, identifies new signatures important for understanding GATOR1 function and its role in cisplatin resistance. Thus, GATOR1 not only participates in the cellular response to amino acid availability but also plays a role in resistance to DNA-damaging anticancer drugs. This novel function of GATOR1 should be taken into account when developing new strategies to combat chemoresistance.

顺铂是许多上皮性癌症的主要化疗方法。然而,对这种药物的耐药性对有效治疗构成了重大挑战。尽管确定了许多与耐药相关的因素,但预测药物反应的可靠生物标志物仍然难以捉摸。此前,NPRL2肿瘤抑制因子的低表达与顺铂耐药有关。NPRL2与NPRL3和DEPDC5一起形成GATOR1复合物,这是mTORС1的上游调节剂,其功能在许多癌症中受到干扰,特别是那些对顺铂具有耐药性的癌症。在这里,我们比较了GATOR1缺失的非癌性支气管上皮BEAS-2B细胞(作为固有顺铂耐药模型)与获得性顺铂耐药的非小细胞肺癌细胞系A549、H460和H1975。我们发现任何GATOR1成员的缺失,而不仅仅是NPRL2,都会促进顺铂耐药性,而它们的过表达会使细胞对药物敏感。在GATOR1缺失的细胞中,顺铂外流所需的ATP7A转运体表达增加,而顺铂内流转运体CTR2和LRRC8A的表达下调,尤其是在用药后。这阻碍了药物在细胞中的积累,导致顺铂- dna加合物的形成减少。同时,这些细胞表现出增强的DNA损伤反应和mTORC1活性。过表达GATOR1组分和/或同时使用mTORC1抑制剂可恢复对顺铂的敏感性。对GATOR1缺失的BEAS-2B细胞进行转录组学分析,发现了对理解GATOR1功能及其在顺铂耐药中的作用很重要的新特征。因此,GATOR1不仅参与细胞对氨基酸可用性的反应,还在抗dna损伤抗癌药物中发挥作用。在制定对抗化疗耐药的新策略时,应考虑到GATOR1的这种新功能。
{"title":"GATOR1 complex controls cisplatin sensitivity.","authors":"Zhenrui Pan, Hanxiao Zhang, Xia Xiao, Catherine Brenner, Svetlana Dokudovskaya","doi":"10.1038/s41419-025-08392-4","DOIUrl":"https://doi.org/10.1038/s41419-025-08392-4","url":null,"abstract":"<p><p>Cisplatin administration is the primary chemotherapy approach for many epithelial cancers. However, resistance to this drug poses a significant challenge to effective treatment. Despite the identification of numerous factors associated with resistance, reliable biomarkers predicting drug response remain elusive. Previously, low expression of the NPRL2 tumor suppressor was linked to cisplatin resistance. NPRL2, along with NPRL3 and DEPDC5, forms the GATOR1 complex, an upstream regulator of the mTORС1, the function of which is perturbed in many cancers, particularly those resistant to cisplatin. Here, we compare non-cancerous bronchial epithelium BEAS-2B cells with GATOR1 deletions, serving as a model of intrinsic cisplatin resistance, with non-small cell lung cancer lines A549, H460, and H1975 with acquired resistance to the drug. We found that deletion of any GATOR1 member, not solely NPRL2, promotes cisplatin resistance, whereas their overexpression renders cells sensitive to the drug. In cells with GATOR1 deletions, expression of the ATP7A transporter required for cisplatin efflux is increased, while expression of cisplatin influx transporters CTR2 and LRRC8A is downregulated, especially after treatment with the drug. This hinders drug accumulation in cells, resulting in the formation of fewer cisplatin-DNA adducts. Simultaneously, these cells exhibit enhanced DNA damage response and mTORC1 activity. Overexpression of GATOR1 components and/or concomitant treatment with an mTORC1 inhibitor restores sensitivity to cisplatin. Transcriptomic analysis of GATOR1-deleted BEAS-2B cells, treated or not with the drug, identifies new signatures important for understanding GATOR1 function and its role in cisplatin resistance. Thus, GATOR1 not only participates in the cellular response to amino acid availability but also plays a role in resistance to DNA-damaging anticancer drugs. This novel function of GATOR1 should be taken into account when developing new strategies to combat chemoresistance.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145862335","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
Unraveling sex differences in age-related hippocampal decline: differential mitochondrial dysfunction, Lonp1-dependent mitochondrial proteostasis and mtROS production in aged C57BL/6 mice. 揭示年龄相关海马衰退的性别差异:老年C57BL/6小鼠线粒体功能障碍、lonp1依赖的线粒体蛋白停滞和mtROS产生的差异。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-30 DOI: 10.1038/s41419-025-08360-y
Karina A Cicali, Claudia Jara, Daniela Cortés-Díaz, Matías Lira, Ítalo Fuentes, Alejandra Catenaccio, Josefa Arnaíz, Micaela Ricca, Sebastián Valenzuela, Carolina A Oliva, Daniela S Rivera, Cheril Tapia-Rojas

Aging is a progressive process characterized by cellular and molecular damage leading to mitochondrial dysfunction and cognitive decline. Mitochondrial dysfunction is a critical factor in memory impairment in aging and neurodegenerative diseases. While sex differences in aging have been observed across various species, the underlying cellular and molecular mechanisms remain poorly understood, mainly focused on mitochondrial proteostasis. This study examined hippocampal-dependent cognitive decline and mitochondrial dysfunction in aged male and female C57BL/6 J mice. Our results reveal sex-dependent differences in cognitive impairment, with aged males exhibiting more significant deficits in spatial and localization memory, while aged females show impairments in recognition memory. Additionally, aged males display increased oxidative stress and exacerbated mitochondrial superoxide production, leading to more severe bioenergetic deficiencies. Conversely, aged females exhibit heightened mitochondrial permeability transition pore (mPTP) activity, suggesting a distinct mechanism of mitochondrial dysfunction, which could explain, almost in part, the cognitive differences in aging. Investigating possible mechanisms responsible for this mitochondrial dysfunction, we found that mitochondrial proteostasis is more prone to failure in aged males, with a significant decrease in the protease activity of Lonp1, a key matrix mitochondrial protease degrading >50% of the mitochondrial proteome. To further reinforce these findings, we replicated key experiments in SAMP8 mice, a model of accelerated aging, obtaining consistent results that strengthen the robustness and generalization of our conclusions. These findings suggest that sex influences hippocampal aging at multiple levels, highlighting the need to consider sexual dimorphism in aging research. This study also emphasizes the critical role of mitochondrial proteostasis in maintaining mitochondrial function in aging in a sex-dependent manner. Understanding these differences could facilitate the development of sex-specific strategies to mitigate age-related cognitive decline and neurodegeneration.

衰老是一个渐进的过程,其特征是细胞和分子损伤导致线粒体功能障碍和认知能力下降。线粒体功能障碍是衰老和神经退行性疾病中记忆障碍的关键因素。虽然在不同的物种中观察到衰老的性别差异,但潜在的细胞和分子机制仍然知之甚少,主要集中在线粒体蛋白质平衡上。本研究研究了老年雄性和雌性C57BL/6 J小鼠海马依赖性认知衰退和线粒体功能障碍。我们的研究结果揭示了认知障碍的性别差异,老年男性在空间和定位记忆方面表现出更明显的缺陷,而老年女性在识别记忆方面表现出更明显的缺陷。此外,老年男性表现出氧化应激增加,线粒体超氧化物产生加剧,导致更严重的生物能量缺乏。相反,老年女性表现出更高的线粒体通透性过渡孔(mPTP)活性,这表明线粒体功能障碍的独特机制,这几乎可以部分解释衰老过程中的认知差异。在研究导致线粒体功能障碍的可能机制时,我们发现老年男性的线粒体蛋白平衡更容易失效,Lonp1蛋白酶活性显著降低,Lonp1是一种关键的基质线粒体蛋白酶,可降解50%的线粒体蛋白质组。为了进一步强化这些发现,我们在加速衰老模型SAMP8小鼠中重复了关键实验,获得了一致的结果,增强了我们结论的稳健性和泛化性。这些发现表明,性别在多个层面上影响海马的衰老,强调了在衰老研究中考虑性别二态性的必要性。这项研究还强调了线粒体蛋白平衡在维持线粒体功能中以性别依赖的方式衰老的关键作用。了解这些差异可以促进性别特异性策略的发展,以减轻与年龄相关的认知能力下降和神经变性。
{"title":"Unraveling sex differences in age-related hippocampal decline: differential mitochondrial dysfunction, Lonp1-dependent mitochondrial proteostasis and mtROS production in aged C57BL/6 mice.","authors":"Karina A Cicali, Claudia Jara, Daniela Cortés-Díaz, Matías Lira, Ítalo Fuentes, Alejandra Catenaccio, Josefa Arnaíz, Micaela Ricca, Sebastián Valenzuela, Carolina A Oliva, Daniela S Rivera, Cheril Tapia-Rojas","doi":"10.1038/s41419-025-08360-y","DOIUrl":"https://doi.org/10.1038/s41419-025-08360-y","url":null,"abstract":"<p><p>Aging is a progressive process characterized by cellular and molecular damage leading to mitochondrial dysfunction and cognitive decline. Mitochondrial dysfunction is a critical factor in memory impairment in aging and neurodegenerative diseases. While sex differences in aging have been observed across various species, the underlying cellular and molecular mechanisms remain poorly understood, mainly focused on mitochondrial proteostasis. This study examined hippocampal-dependent cognitive decline and mitochondrial dysfunction in aged male and female C57BL/6 J mice. Our results reveal sex-dependent differences in cognitive impairment, with aged males exhibiting more significant deficits in spatial and localization memory, while aged females show impairments in recognition memory. Additionally, aged males display increased oxidative stress and exacerbated mitochondrial superoxide production, leading to more severe bioenergetic deficiencies. Conversely, aged females exhibit heightened mitochondrial permeability transition pore (mPTP) activity, suggesting a distinct mechanism of mitochondrial dysfunction, which could explain, almost in part, the cognitive differences in aging. Investigating possible mechanisms responsible for this mitochondrial dysfunction, we found that mitochondrial proteostasis is more prone to failure in aged males, with a significant decrease in the protease activity of Lonp1, a key matrix mitochondrial protease degrading >50% of the mitochondrial proteome. To further reinforce these findings, we replicated key experiments in SAMP8 mice, a model of accelerated aging, obtaining consistent results that strengthen the robustness and generalization of our conclusions. These findings suggest that sex influences hippocampal aging at multiple levels, highlighting the need to consider sexual dimorphism in aging research. This study also emphasizes the critical role of mitochondrial proteostasis in maintaining mitochondrial function in aging in a sex-dependent manner. Understanding these differences could facilitate the development of sex-specific strategies to mitigate age-related cognitive decline and neurodegeneration.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145862474","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
Fbxo2 suppresses prostate cancer progression by regulating YTHDF2 ubiquitination and degradation. Fbxo2通过调节YTHDF2泛素化和降解抑制前列腺癌进展。
IF 9.6 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2025-12-29 DOI: 10.1038/s41419-025-08396-0
Xinyu Xu, Guangcheng Dai, Chun-Ling Liu, Qiu Yao, Xiaowei Cai, Yang Wang, Zeyu Chen, Kang Liu, Jin Zhu, Jia Ma, Zhiwei Wang, Boxin Xue, Lixia Wang

Deregulation of E3 ubiquitin ligases is associated with increased proliferation and metastasis in prostate cancer (PCa); however, the underlying mechanisms remain largely unclear. This study aimed to explore the role of Fbxo2, a SKP1-Cullin-F-box (SCF) E3 ubiquitin ligase, in PCa progression. Analysis of prostate tissue samples revealed that Fbxo2 is downregulated in PCa, and higher Fbxo2 expression correlates with better patient prognosis. Functional assays conducted both in vitro and in vivo demonstrated that Fbxo2 reduces cell proliferation and metastasis in PCa. Using co-immunoprecipitation mass spectrometry (co-IP-MS), co-IP, western blotting, and ubiquitin assays, we identified that m6A reader YTHDF2, an oncoprotein that is upregulated in PCa, was a substrate of Fbxo2-mediated degradation. Notably, Fbxo2 mutants lacking the C-terminal region were less effective in promoting YTHDF2 ubiquitination and destruction. Furthermore, lysine 286 (K286) of YTHDF2 was identified as the key ubiquitination site. A series of rescue experiments revealed that silencing or overexpressing YTHDF2 modulated the effects of Fbxo2 knockdown or overexpression, confirming their functional interplay. Mechanistically, YTHDF2 enhanced the PCa progression and metastasis by modulating the m6A methylation of CDKN1C mRNA. Together, these findings suggest that Fbxo2 axis may serve as a potential prognostic marker and therapeutic target in PCa.

E3泛素连接酶的解除与前列腺癌(PCa)的增殖和转移增加有关;然而,潜在的机制在很大程度上仍不清楚。本研究旨在探讨SKP1-Cullin-F-box (SCF) E3泛素连接酶Fbxo2在PCa进展中的作用。前列腺组织样本分析显示,Fbxo2在前列腺癌中表达下调,Fbxo2表达越高,患者预后越好。体外和体内的功能分析表明,Fbxo2可以降低前列腺癌细胞的增殖和转移。通过共免疫沉淀质谱(co-IP- ms)、共ip、western blotting和泛素测定,我们发现m6A读取器YTHDF2是一种在PCa中上调的癌蛋白,是fbxo2介导的降解的底物。值得注意的是,缺乏c端区域的Fbxo2突变体在促进YTHDF2泛素化和破坏方面效果较差。此外,YTHDF2的赖氨酸286 (K286)被确定为关键的泛素化位点。一系列修复实验显示,沉默或过表达YTHDF2可调节Fbxo2敲低或过表达的作用,证实了它们在功能上的相互作用。在机制上,YTHDF2通过调节CDKN1C mRNA的m6A甲基化来促进前列腺癌的进展和转移。总之,这些发现表明Fbxo2轴可能作为前列腺癌的潜在预后标志物和治疗靶点。
{"title":"Fbxo2 suppresses prostate cancer progression by regulating YTHDF2 ubiquitination and degradation.","authors":"Xinyu Xu, Guangcheng Dai, Chun-Ling Liu, Qiu Yao, Xiaowei Cai, Yang Wang, Zeyu Chen, Kang Liu, Jin Zhu, Jia Ma, Zhiwei Wang, Boxin Xue, Lixia Wang","doi":"10.1038/s41419-025-08396-0","DOIUrl":"https://doi.org/10.1038/s41419-025-08396-0","url":null,"abstract":"<p><p>Deregulation of E3 ubiquitin ligases is associated with increased proliferation and metastasis in prostate cancer (PCa); however, the underlying mechanisms remain largely unclear. This study aimed to explore the role of Fbxo2, a SKP1-Cullin-F-box (SCF) E3 ubiquitin ligase, in PCa progression. Analysis of prostate tissue samples revealed that Fbxo2 is downregulated in PCa, and higher Fbxo2 expression correlates with better patient prognosis. Functional assays conducted both in vitro and in vivo demonstrated that Fbxo2 reduces cell proliferation and metastasis in PCa. Using co-immunoprecipitation mass spectrometry (co-IP-MS), co-IP, western blotting, and ubiquitin assays, we identified that m6A reader YTHDF2, an oncoprotein that is upregulated in PCa, was a substrate of Fbxo2-mediated degradation. Notably, Fbxo2 mutants lacking the C-terminal region were less effective in promoting YTHDF2 ubiquitination and destruction. Furthermore, lysine 286 (K286) of YTHDF2 was identified as the key ubiquitination site. A series of rescue experiments revealed that silencing or overexpressing YTHDF2 modulated the effects of Fbxo2 knockdown or overexpression, confirming their functional interplay. Mechanistically, YTHDF2 enhanced the PCa progression and metastasis by modulating the m6A methylation of CDKN1C mRNA. Together, these findings suggest that Fbxo2 axis may serve as a potential prognostic marker and therapeutic target in PCa.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":" ","pages":""},"PeriodicalIF":9.6,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145854703","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 & Disease
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