首页 > 最新文献

Cell Death & Disease最新文献

英文 中文
SMAD4 Limits PARP1 dependent DNA Repair to Render Pancreatic Cancer Cells Sensitive to Radiotherapy. SMAD4 限制 PARP1 依赖性 DNA 修复,使胰腺癌细胞对放疗敏感。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-11 DOI: 10.1038/s41419-024-07210-7
Yang Wang, Tianyu Yu, Zhangting Zhao, Xiaobing Li, Yiran Song, Yazhi He, Yingqun Zhou, Pu Li, Liwei An, Feng Wang

Dysregulation of SMAD4 (i.e. somatic mutation) is strongly associated with poor pancreatic ductal adenocarcinoma (PDAC) prognosis, yet the molecular mechanisms remain underlying this relationship obscure. Previously, we discovered that SMAD4 mutation renders pancreatic cancer resistant to radiotherapy via promotion of autophagy. In the current work, we observed a downregulation of the protein level of SMAD4 in PDAC as compared with adjacent normal tissue, and that such SMAD4low PDAC failed to benefit from chemotherapy. Furthermore, we observed that SMAD4 depletion dramatically enhanced DNA repair capacity in response to irradiation (IR) or a radiomimetic chemical. Interestingly, we found the radiomimetic chemical having induced a robust translocation of SMAD4 into the nucleus, where a direct interaction was shown to occur between the MH1 domain of SMAD4 and the DBD domain of PARP1. Functionally, the SMAD4-PARP1 interaction was found to perturb the recruitment of PARP1 to DNA damage sites. Accordingly, the combination of olaparib and radiotherapy was indicated in vivo and in vitro to specifically reduce the growth of SMAD4-deficient PDAC by attenuating PARP1 activity. Collectively, our results revealed a novel molecular mechanism for the involvement of the SMAD4-PARP1 interaction in DNA repair with a vital role in radiotherapy response in PDAC. Based on our set of findings, our findings offer a new combined therapeutic strategy for SMAD4 deficient PDAC that can significantly reduce pancreatic cancer radiotherapy resistance.

SMAD4的失调(即体细胞突变)与胰腺导管腺癌(PDAC)的不良预后密切相关,但这一关系的分子机制仍然模糊不清。此前,我们发现 SMAD4 突变通过促进自噬使胰腺癌对放疗产生耐药性。在目前的研究中,我们观察到与邻近的正常组织相比,SMAD4 在 PDAC 中的蛋白水平下调,而且这种 SMAD4 低的 PDAC 无法从化疗中获益。此外,我们还观察到,SMAD4 的缺失显著增强了对辐照(IR)或辐射模拟化学物质的 DNA 修复能力。有趣的是,我们发现放射性模拟化学物质诱导了SMAD4向细胞核内的稳健转位,在那里,SMAD4的MH1结构域和PARP1的DBD结构域之间发生了直接的相互作用。从功能上讲,SMAD4-PARP1 的相互作用会扰乱 PARP1 在 DNA 损伤位点的招募。因此,奥拉帕利和放疗的体内和体外联合应用表明,通过削弱PARP1的活性,可以特异性地减少SMAD4缺陷型PDAC的生长。总之,我们的研究结果揭示了一种新的分子机制,即SMAD4-PARP1相互作用参与DNA修复,并在PDAC的放疗反应中发挥重要作用。基于我们的一系列发现,我们的研究结果为SMAD4缺陷型PDAC提供了一种新的联合治疗策略,可显著降低胰腺癌放疗耐药性。
{"title":"SMAD4 Limits PARP1 dependent DNA Repair to Render Pancreatic Cancer Cells Sensitive to Radiotherapy.","authors":"Yang Wang, Tianyu Yu, Zhangting Zhao, Xiaobing Li, Yiran Song, Yazhi He, Yingqun Zhou, Pu Li, Liwei An, Feng Wang","doi":"10.1038/s41419-024-07210-7","DOIUrl":"10.1038/s41419-024-07210-7","url":null,"abstract":"<p><p>Dysregulation of SMAD4 (i.e. somatic mutation) is strongly associated with poor pancreatic ductal adenocarcinoma (PDAC) prognosis, yet the molecular mechanisms remain underlying this relationship obscure. Previously, we discovered that SMAD4 mutation renders pancreatic cancer resistant to radiotherapy via promotion of autophagy. In the current work, we observed a downregulation of the protein level of SMAD4 in PDAC as compared with adjacent normal tissue, and that such SMAD4<sup>low</sup> PDAC failed to benefit from chemotherapy. Furthermore, we observed that SMAD4 depletion dramatically enhanced DNA repair capacity in response to irradiation (IR) or a radiomimetic chemical. Interestingly, we found the radiomimetic chemical having induced a robust translocation of SMAD4 into the nucleus, where a direct interaction was shown to occur between the MH1 domain of SMAD4 and the DBD domain of PARP1. Functionally, the SMAD4-PARP1 interaction was found to perturb the recruitment of PARP1 to DNA damage sites. Accordingly, the combination of olaparib and radiotherapy was indicated in vivo and in vitro to specifically reduce the growth of SMAD4-deficient PDAC by attenuating PARP1 activity. Collectively, our results revealed a novel molecular mechanism for the involvement of the SMAD4-PARP1 interaction in DNA repair with a vital role in radiotherapy response in PDAC. Based on our set of findings, our findings offer a new combined therapeutic strategy for SMAD4 deficient PDAC that can significantly reduce pancreatic cancer radiotherapy resistance.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"818"},"PeriodicalIF":8.1,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555233/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615571","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PRMT1-mediated methylation of ME2 promotes hepatocellular carcinoma growth by inhibiting ubiquitination. PRMT1 介导的 ME2 甲基化通过抑制泛素化促进肝细胞癌的生长。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-11 DOI: 10.1038/s41419-024-07219-y
Shuai Zhang, Shuling Zhang, Baijuan Xia, Xueying Li, Hongyu Jiang, Su Feng, Yang Xiang, Ya Qiu, Shi Zhou, Peng Luo

The mitochondrial malic enzyme 2 (ME2), which is frequently elevated during carcinogenesis and may be a target for cancer therapy, catalyzes the conversion of malate to pyruvate. The processes controlling ME2 activity, however, remain largely unclear. In this work, we show that human hepatocellular carcinoma (HCC) tissues contain high levels of ME2 and that the methylation of ME2 stimulates the growth and migration of HCC cells. Furthermore, we observed that ME2 interacts with protein arginine methyltransferase 1 (PRMT1) and that ME2 enzymatic activity is activated by mutation of ME2 at lysine 67. Mitochondrial respiration was markedly increased by activated ME2, which promoted cell division and carcinogenesis. Furthermore, a negative prognosis for patients was strongly linked with the expression levels of PRMT1 and ME2 R67K in HCC tissues. These findings imply that hepatocellular carcinoma growth is aided by PRMT1-mediated ME2 methylation, that is an essential signaling event that cancer cells need to continue mitochondrial respiration.

线粒体苹果酸酶 2(ME2)催化苹果酸转化为丙酮酸,它在癌变过程中经常升高,可能成为癌症治疗的靶点。然而,控制 ME2 活性的过程在很大程度上仍不清楚。在这项研究中,我们发现人类肝细胞癌(HCC)组织中含有大量 ME2,而且 ME2 的甲基化会刺激 HCC 细胞的生长和迁移。此外,我们还观察到 ME2 与蛋白精氨酸甲基转移酶 1(PRMT1)相互作用,ME2 的酶活性可通过 ME2 在赖氨酸 67 处的突变被激活。活化的 ME2 能显著增加线粒体呼吸,促进细胞分裂和癌变。此外,患者的不良预后与肝癌组织中 PRMT1 和 ME2 R67K 的表达水平密切相关。这些研究结果表明,PRMT1介导的ME2甲基化有助于肝细胞癌的生长,而ME2甲基化是癌细胞继续进行线粒体呼吸所必需的信号转导。
{"title":"PRMT1-mediated methylation of ME2 promotes hepatocellular carcinoma growth by inhibiting ubiquitination.","authors":"Shuai Zhang, Shuling Zhang, Baijuan Xia, Xueying Li, Hongyu Jiang, Su Feng, Yang Xiang, Ya Qiu, Shi Zhou, Peng Luo","doi":"10.1038/s41419-024-07219-y","DOIUrl":"10.1038/s41419-024-07219-y","url":null,"abstract":"<p><p>The mitochondrial malic enzyme 2 (ME2), which is frequently elevated during carcinogenesis and may be a target for cancer therapy, catalyzes the conversion of malate to pyruvate. The processes controlling ME2 activity, however, remain largely unclear. In this work, we show that human hepatocellular carcinoma (HCC) tissues contain high levels of ME2 and that the methylation of ME2 stimulates the growth and migration of HCC cells. Furthermore, we observed that ME2 interacts with protein arginine methyltransferase 1 (PRMT1) and that ME2 enzymatic activity is activated by mutation of ME2 at lysine 67. Mitochondrial respiration was markedly increased by activated ME2, which promoted cell division and carcinogenesis. Furthermore, a negative prognosis for patients was strongly linked with the expression levels of PRMT1 and ME2 R67K in HCC tissues. These findings imply that hepatocellular carcinoma growth is aided by PRMT1-mediated ME2 methylation, that is an essential signaling event that cancer cells need to continue mitochondrial respiration.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"814"},"PeriodicalIF":8.1,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555414/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615634","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Single-cell landscape identified SERPINB9 as a key player contributing to stemness and metastasis in non-seminomas. 单细胞图谱发现,SERPINB9是导致非肿瘤干性和转移的关键因素。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-11 DOI: 10.1038/s41419-024-07220-5
Zhouliang Bian, Biying Chen, Guohai Shi, Haihua Yuan, Yue Zhou, Bin Jiang, Long Li, Hengchuan Su, Yanjie Zhang

Embryonal carcinoma (EC), characterized by a high degree of stemness similar to that of embryonic stem cells, is the most malignant subtype within non-seminomatous testicular germ cell tumors (TGCTs). However, the mechanisms underlying its malignancy remain unknown. In this study, we employed single-cell RNA sequencing to analyze four non-seminoma samples. Our differential expression analysis revealed high expression of SERPINB9 in metastatic EC cells. We conducted in vitro experiments to further investigate SERPINB9's role in the progression of EC. Functionally, the knockdown of SERPINB9 in NCCIT and NTERA-2 leads to a diminished migratory capability and decreased cis-platin resistance, as demonstrated by Transwell migration assay and drug sensitivity assay. Moreover, embryoid bodies showed reduced size and lower OCT4 expression, alongside heightened expression of differentiation markers AFP, ACTA2, and CD57 in shSERPINB9 cells. In vivo, the role of SERPINB9 in maintaining cancer stemness was validated by the limiting dilution assay. Mechanistically, Bulk RNA-seq further showed downregulation of ERK1/2 signaling and WNT signaling pathways with concomitant upregulation of differentiation pathways subsequent to SERPINB9 knockdown. Additionally, the analysis indicated increased levels of cytokines linked to tertiary lymphoid structures (TLS), such as IL6, IL11, IL15, CCL2, CCL5, and CXCL13 in shSERPINB9 cells, which were further validated by ELISA. Our research indicates that SERPINB9 plays a key role in driving tumor progression by enhancing tumor stemness and suppressing TLS. This study stands as the first to elucidate the molecular signature of non-seminomas at a single-cell level, presenting a wealth of promising targets with substantial potential for informing the development of future therapeutic interventions.

胚胎干细胞癌(EC)具有类似胚胎干细胞的高度干性,是非肉芽肿性睾丸生殖细胞肿瘤(TGCT)中恶性程度最高的亚型。然而,其恶性机制仍不为人知。在这项研究中,我们采用单细胞 RNA 测序分析了四个非精原细胞瘤样本。我们的差异表达分析表明,SERPINB9在转移性EC细胞中高表达。我们进行了体外实验,进一步研究SERPINB9在EC进展过程中的作用。从功能上讲,在NCCIT和NTERA-2中敲除SERPINB9会导致迁移能力减弱和顺铂抗性降低,这一点已在Transwell迁移试验和药物敏感性试验中得到证实。此外,在 shSERPINB9 细胞中,胚状体的大小减小,OCT4 表达降低,分化标志物 AFP、ACTA2 和 CD57 的表达增加。在体内,SERPINB9在维持癌症干性方面的作用得到了限制稀释试验的验证。从机理上讲,大量 RNA-seq 进一步表明,SERPINB9 基因敲除后,ERK1/2 信号通路和 WNT 信号通路下调,同时分化通路上调。此外,分析表明,在 shSERPINB9 细胞中,与三级淋巴结构(TLS)相关的细胞因子(如 IL6、IL11、IL15、CCL2、CCL5 和 CXCL13)水平升高,ELISA 进一步验证了这一点。我们的研究表明,SERPINB9 通过增强肿瘤干性和抑制 TLS 在推动肿瘤进展中发挥着关键作用。这项研究首次在单细胞水平上阐明了非骨髓瘤的分子特征,为未来治疗干预措施的开发提供了大量有潜力的靶点。
{"title":"Single-cell landscape identified SERPINB9 as a key player contributing to stemness and metastasis in non-seminomas.","authors":"Zhouliang Bian, Biying Chen, Guohai Shi, Haihua Yuan, Yue Zhou, Bin Jiang, Long Li, Hengchuan Su, Yanjie Zhang","doi":"10.1038/s41419-024-07220-5","DOIUrl":"10.1038/s41419-024-07220-5","url":null,"abstract":"<p><p>Embryonal carcinoma (EC), characterized by a high degree of stemness similar to that of embryonic stem cells, is the most malignant subtype within non-seminomatous testicular germ cell tumors (TGCTs). However, the mechanisms underlying its malignancy remain unknown. In this study, we employed single-cell RNA sequencing to analyze four non-seminoma samples. Our differential expression analysis revealed high expression of SERPINB9 in metastatic EC cells. We conducted in vitro experiments to further investigate SERPINB9's role in the progression of EC. Functionally, the knockdown of SERPINB9 in NCCIT and NTERA-2 leads to a diminished migratory capability and decreased cis-platin resistance, as demonstrated by Transwell migration assay and drug sensitivity assay. Moreover, embryoid bodies showed reduced size and lower OCT4 expression, alongside heightened expression of differentiation markers AFP, ACTA2, and CD57 in shSERPINB9 cells. In vivo, the role of SERPINB9 in maintaining cancer stemness was validated by the limiting dilution assay. Mechanistically, Bulk RNA-seq further showed downregulation of ERK1/2 signaling and WNT signaling pathways with concomitant upregulation of differentiation pathways subsequent to SERPINB9 knockdown. Additionally, the analysis indicated increased levels of cytokines linked to tertiary lymphoid structures (TLS), such as IL6, IL11, IL15, CCL2, CCL5, and CXCL13 in shSERPINB9 cells, which were further validated by ELISA. Our research indicates that SERPINB9 plays a key role in driving tumor progression by enhancing tumor stemness and suppressing TLS. This study stands as the first to elucidate the molecular signature of non-seminomas at a single-cell level, presenting a wealth of promising targets with substantial potential for informing the development of future therapeutic interventions.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"812"},"PeriodicalIF":8.1,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555415/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615570","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inflammation in atherosclerosis: pathophysiology and mechanisms. 动脉粥样硬化中的炎症:病理生理学和机制。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-11 DOI: 10.1038/s41419-024-07166-8
Amir Ajoolabady, Domenico Pratico, Ling Lin, Christos S Mantzoros, Suhad Bahijri, Jaakko Tuomilehto, Jun Ren

Atherosclerosis imposes a heavy burden on cardiovascular health due to its indispensable role in the pathogenesis of cardiovascular disease (CVD) such as coronary artery disease and heart failure. Ample clinical and experimental evidence has corroborated the vital role of inflammation in the pathophysiology of atherosclerosis. Hence, the demand for preclinical research into atherosclerotic inflammation is on the horizon. Indeed, the acquisition of an in-depth knowledge of the molecular and cellular mechanisms of inflammation in atherosclerosis should allow us to identify novel therapeutic targets with translational merits. In this review, we aimed to critically discuss and speculate on the recently identified molecular and cellular mechanisms of inflammation in atherosclerosis. Moreover, we delineated various signaling cascades and proinflammatory responses in macrophages and other leukocytes that promote plaque inflammation and atherosclerosis. In the end, we highlighted potential therapeutic targets, the pros and cons of current interventions, as well as anti-inflammatory and atheroprotective mechanisms.

动脉粥样硬化在冠心病和心力衰竭等心血管疾病(CVD)的发病机制中扮演着不可或缺的角色,给心血管健康带来了沉重负担。大量临床和实验证据证实了炎症在动脉粥样硬化病理生理学中的重要作用。因此,对动脉粥样硬化炎症的临床前研究的需求即将到来。事实上,深入了解动脉粥样硬化中炎症的分子和细胞机制,可以让我们找到具有转化价值的新型治疗靶点。在这篇综述中,我们旨在对最近发现的动脉粥样硬化的分子和细胞炎症机制进行批判性讨论和推测。此外,我们还描述了巨噬细胞和其他白细胞中促进斑块炎症和动脉粥样硬化的各种信号级联和促炎反应。最后,我们强调了潜在的治疗目标、当前干预措施的利弊以及抗炎和动脉粥样硬化保护机制。
{"title":"Inflammation in atherosclerosis: pathophysiology and mechanisms.","authors":"Amir Ajoolabady, Domenico Pratico, Ling Lin, Christos S Mantzoros, Suhad Bahijri, Jaakko Tuomilehto, Jun Ren","doi":"10.1038/s41419-024-07166-8","DOIUrl":"10.1038/s41419-024-07166-8","url":null,"abstract":"<p><p>Atherosclerosis imposes a heavy burden on cardiovascular health due to its indispensable role in the pathogenesis of cardiovascular disease (CVD) such as coronary artery disease and heart failure. Ample clinical and experimental evidence has corroborated the vital role of inflammation in the pathophysiology of atherosclerosis. Hence, the demand for preclinical research into atherosclerotic inflammation is on the horizon. Indeed, the acquisition of an in-depth knowledge of the molecular and cellular mechanisms of inflammation in atherosclerosis should allow us to identify novel therapeutic targets with translational merits. In this review, we aimed to critically discuss and speculate on the recently identified molecular and cellular mechanisms of inflammation in atherosclerosis. Moreover, we delineated various signaling cascades and proinflammatory responses in macrophages and other leukocytes that promote plaque inflammation and atherosclerosis. In the end, we highlighted potential therapeutic targets, the pros and cons of current interventions, as well as anti-inflammatory and atheroprotective mechanisms.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"817"},"PeriodicalIF":8.1,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555284/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615566","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting G-quadruplex by TMPyP4 for inhibition of colorectal cancer through cell cycle arrest and boosting anti-tumor immunity. 利用 TMPyP4 靶向 G-四叠体,通过细胞周期停滞抑制结直肠癌并增强抗肿瘤免疫力。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-11 DOI: 10.1038/s41419-024-07215-2
Peisi Li, Dawang Zhou, Yumo Xie, Ze Yuan, Mingzhe Huang, Gaopo Xu, Junfeng Huang, Zhuokai Zhuang, Yanxin Luo, Huichuan Yu, Xiaolin Wang

G-quadruplex (G4) is a noncanonical DNA secondary structure known to induce DNA damage and regulate the expression of immune-related genes. We aim to exploit the G4 folding as a treatment strategy to trigger anti-tumor immune response. In this study, we observe that the abundant genomic G4 in epithelial cells coexists with increased infiltration of CD8+ T cells in colorectal cancer tissue. Furthermore, our data substantiate the inhibitory effect of the G4 ligand TMPyP4 on cancer progression while concurrently stimulating anti-tumor immunity. Mechanistically, TMPyP4 impedes cancer cell proliferation and induces G2/M cell cycle arrest. Additionally, in vivo experiments demonstrate that TMPyP4 enhances the anti-tumor immune response by triggering DNA damage and activating the cGAS-STING pathway, which fosters CD8+ T cell activation and dendritic cell maturation. Importantly, the combined treatment of TMPyP4 and anti-PD1 exhibits a synergistic therapeutic effect on colorectal cancer. In summary, our findings underscore the potential of the G4 ligand TMPyP4 as a dual strategy to target colorectal cancer: inhibiting cancer progression and augmenting anti-tumor immunity through the activation of cGAS-STING pathway.

G-quadruplex (G4) 是一种非规范 DNA 二级结构,已知可诱导 DNA 损伤并调节免疫相关基因的表达。我们旨在利用 G4 折叠作为一种治疗策略,以触发抗肿瘤免疫反应。在这项研究中,我们观察到上皮细胞中丰富的基因组 G4 与结直肠癌组织中 CD8+ T 细胞浸润的增加同时存在。此外,我们的数据证实了 G4 配体 TMPyP4 对癌症进展的抑制作用,同时还能刺激抗肿瘤免疫。从机理上讲,TMPyP4 可阻碍癌细胞增殖并诱导 G2/M 细胞周期停滞。此外,体内实验证明,TMPyP4 通过引发 DNA 损伤和激活 cGAS-STING 通路,促进 CD8+ T 细胞活化和树突状细胞成熟,从而增强抗肿瘤免疫反应。重要的是,TMPyP4 和抗 PD1 的联合治疗对结直肠癌具有协同治疗效果。总之,我们的研究结果强调了 G4 配体 TMPyP4 作为针对结直肠癌的双重策略的潜力:通过激活 cGAS-STING 通路抑制癌症进展和增强抗肿瘤免疫力。
{"title":"Targeting G-quadruplex by TMPyP4 for inhibition of colorectal cancer through cell cycle arrest and boosting anti-tumor immunity.","authors":"Peisi Li, Dawang Zhou, Yumo Xie, Ze Yuan, Mingzhe Huang, Gaopo Xu, Junfeng Huang, Zhuokai Zhuang, Yanxin Luo, Huichuan Yu, Xiaolin Wang","doi":"10.1038/s41419-024-07215-2","DOIUrl":"10.1038/s41419-024-07215-2","url":null,"abstract":"<p><p>G-quadruplex (G4) is a noncanonical DNA secondary structure known to induce DNA damage and regulate the expression of immune-related genes. We aim to exploit the G4 folding as a treatment strategy to trigger anti-tumor immune response. In this study, we observe that the abundant genomic G4 in epithelial cells coexists with increased infiltration of CD8<sup>+</sup> T cells in colorectal cancer tissue. Furthermore, our data substantiate the inhibitory effect of the G4 ligand TMPyP4 on cancer progression while concurrently stimulating anti-tumor immunity. Mechanistically, TMPyP4 impedes cancer cell proliferation and induces G2/M cell cycle arrest. Additionally, in vivo experiments demonstrate that TMPyP4 enhances the anti-tumor immune response by triggering DNA damage and activating the cGAS-STING pathway, which fosters CD8<sup>+</sup> T cell activation and dendritic cell maturation. Importantly, the combined treatment of TMPyP4 and anti-PD1 exhibits a synergistic therapeutic effect on colorectal cancer. In summary, our findings underscore the potential of the G4 ligand TMPyP4 as a dual strategy to target colorectal cancer: inhibiting cancer progression and augmenting anti-tumor immunity through the activation of cGAS-STING pathway.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"816"},"PeriodicalIF":8.1,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11554887/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142635917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GNG5 is a novel regulator of Aβ42 production in Alzheimer's disease. GNG5 是阿尔茨海默病中 Aβ42 生成的新型调节因子。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-11 DOI: 10.1038/s41419-024-07218-z
Chunyuan Li, Yan Yang, Shiqi Luo, Wenying Qiu, Xia Wang, Wei Ge

The therapeutic options for Alzheimer's disease (AD) are limited, underscoring the critical need for finding an effective regulator of Aβ42 production. In this study, with 489 human postmortem brains, we revealed that homotrimer G protein subunit gamma 5 (GNG5) expression is upregulated in the hippocampal-entorhinal region of pathological AD compared with normal controls, and is positively correlated with Aβ pathology. In vivo and in vitro experiments confirm that increased GNG5 significantly promotes Aβ pathology and Aβ42 production. Mechanically, GNG5 regulates the cleavage preference of γ-secretase towards Aβ42 by directly interacting with the γ-secretase catalytic subunit presenilin 1 (PS1). Moreover, excessive GNG5 increases the protein levels and the activation of Rab5, leading to the increased number of early endosomes, the major cellular organelle for production of Aβ42. Furthermore, immunoprecipitation and immunofluorescence revealed co-interaction of Aβ42 with GPCR family CXCR2, which is known as the receptor for IL-8, thus facilitating the dissociation of G-proteins βγ from α subunits. Treatment of Aβ42 in neurons combined with structure prediction indicated Aβ42 oligomers as a new ligand of CXCR2, upregulating γ subunit GNG5 protein levels. The co-localizations of GNG5 and PS1, CXCR2 and Aβ42 were verified in eight human brain regions. Besides, GNG5 is significantly reduced in extracellular vesicles (EVs) derived from cerebral cortex or serum of AD patients compared with healthy cognition controls. In brief, GNG5 is a novel regulator of Aβ42 production, suggesting its clinical potential as a diagnosis biomarker and the therapeutic target for AD. The GNG5 content in EVs derived from serum and brain tissue of patients with AD significantly reduced. The GNG5 expression in the hippocampal-entorhinal neurons of donors with pathological AD significantly increased, and can exist in homotrimer subtypes. GNG5 expression positively correlates with Aβ pathology and Aβ42 production. Homotrimer-GNG5 binds to the γ-secretase catalytic subunit PS1 and preferentially generates Aβ42 in early endosome. GNG5 leads to enhanced Rab5 protein and activation levels, increased number of early endosome, promoting Aβ42 production. Further, Aβ42 binds to CXCR2 to upregulate GNG5 levels in a feedback loop.

阿尔茨海默病(AD)的治疗方案很有限,因此迫切需要找到一种有效的 Aβ42 生成调节剂。在这项研究中,我们利用 489 个人类死后大脑发现,与正常对照组相比,同三聚体 G 蛋白亚基γ 5(GNG5)在病理性 AD 的海马-脑室区域表达上调,并且与 Aβ 的病理变化呈正相关。体内和体外实验证实,GNG5 的增加会显著促进 Aβ 的病理变化和 Aβ42 的产生。在机制上,GNG5 通过直接与γ-分泌酶催化亚基 presenilin 1(PS1)相互作用,调节γ-分泌酶对 Aβ42 的裂解偏好。此外,过量的 GNG5 会增加 Rab5 的蛋白水平并激活 Rab5,导致早期内体(产生 Aβ42 的主要细胞器)数量增加。此外,免疫沉淀和免疫荧光显示 Aβ42 与 GPCR 家族 CXCR2(众所周知的 IL-8 受体)共同作用,从而促进 G 蛋白 βγ 与 α 亚基的解离。在神经元中处理 Aβ42 并进行结构预测表明,Aβ42 寡聚体是 CXCR2 的新配体,能上调 γ 亚基 GNG5 蛋白水平。在八个人脑区域中,GNG5与PS1、CXCR2和Aβ42的共定位得到了验证。此外,与健康认知对照组相比,从AD患者大脑皮层或血清中提取的细胞外囊泡(EVs)中的GNG5明显减少。简而言之,GNG5是Aβ42生成的新型调节因子,表明它具有作为AD诊断生物标志物和治疗靶点的临床潜力。从AD患者血清和脑组织中提取的EVs中的GNG5含量明显降低。病理AD供体的海马-脑干神经元中GNG5的表达量明显增加,并可存在于同源三聚体亚型中。GNG5的表达与Aβ病理学和Aβ42的产生呈正相关。同源三聚体-GNG5与γ-分泌酶催化亚基PS1结合,在早期内质体中优先生成Aβ42。GNG5 导致 Rab5 蛋白和活化水平提高,早期内质体数量增加,促进了 Aβ42 的生成。此外,Aβ42 与 CXCR2 结合,在反馈环中上调 GNG5 的水平。
{"title":"GNG5 is a novel regulator of Aβ42 production in Alzheimer's disease.","authors":"Chunyuan Li, Yan Yang, Shiqi Luo, Wenying Qiu, Xia Wang, Wei Ge","doi":"10.1038/s41419-024-07218-z","DOIUrl":"10.1038/s41419-024-07218-z","url":null,"abstract":"<p><p>The therapeutic options for Alzheimer's disease (AD) are limited, underscoring the critical need for finding an effective regulator of Aβ42 production. In this study, with 489 human postmortem brains, we revealed that homotrimer G protein subunit gamma 5 (GNG5) expression is upregulated in the hippocampal-entorhinal region of pathological AD compared with normal controls, and is positively correlated with Aβ pathology. In vivo and in vitro experiments confirm that increased GNG5 significantly promotes Aβ pathology and Aβ42 production. Mechanically, GNG5 regulates the cleavage preference of γ-secretase towards Aβ42 by directly interacting with the γ-secretase catalytic subunit presenilin 1 (PS1). Moreover, excessive GNG5 increases the protein levels and the activation of Rab5, leading to the increased number of early endosomes, the major cellular organelle for production of Aβ42. Furthermore, immunoprecipitation and immunofluorescence revealed co-interaction of Aβ42 with GPCR family CXCR2, which is known as the receptor for IL-8, thus facilitating the dissociation of G-proteins βγ from α subunits. Treatment of Aβ42 in neurons combined with structure prediction indicated Aβ42 oligomers as a new ligand of CXCR2, upregulating γ subunit GNG5 protein levels. The co-localizations of GNG5 and PS1, CXCR2 and Aβ42 were verified in eight human brain regions. Besides, GNG5 is significantly reduced in extracellular vesicles (EVs) derived from cerebral cortex or serum of AD patients compared with healthy cognition controls. In brief, GNG5 is a novel regulator of Aβ42 production, suggesting its clinical potential as a diagnosis biomarker and the therapeutic target for AD. The GNG5 content in EVs derived from serum and brain tissue of patients with AD significantly reduced. The GNG5 expression in the hippocampal-entorhinal neurons of donors with pathological AD significantly increased, and can exist in homotrimer subtypes. GNG5 expression positively correlates with Aβ pathology and Aβ42 production. Homotrimer-GNG5 binds to the γ-secretase catalytic subunit PS1 and preferentially generates Aβ42 in early endosome. GNG5 leads to enhanced Rab5 protein and activation levels, increased number of early endosome, promoting Aβ42 production. Further, Aβ42 binds to CXCR2 to upregulate GNG5 levels in a feedback loop.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"815"},"PeriodicalIF":8.1,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11554683/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615561","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Combined metformin and simvastatin therapy inhibits SREBP2 maturation and alters energy metabolism in glioma. 二甲双胍和辛伐他汀联合疗法可抑制胶质瘤中SREBP2的成熟并改变其能量代谢。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-09 DOI: 10.1038/s41419-024-07169-5
Xiaolong Qiao, Zixuan Wang, Yinan Chen, Nan Peng, Hongwei Zhang, Chaoshi Niu, Chuandong Cheng

This study aims to explore the inhibitory effects of combined metformin and simvastatin therapy on the malignant progression of glioma. The research specifically examines how the maturation of SREBP2 as a transcription factor affects the expression of GLUT1 and GLUT6 in glioma cells. Additionally, it investigates the impact of this combination therapy on the biological functions and energy metabolism of glioma cells. To assess the functions of GLUT1/6, sh-GLUT1/6 plasmids were employed. The study determined the half-maximal inhibitory concentrations (IC50) of metformin and simvastatin using the CCK-8 assay. Subsequently, the effects of these drugs on glioma metabolism, proliferation, and apoptosis were explored in vitro and in vivo, using drug concentrations significantly lower than their respective IC50 values. The impact of drug treatment on GLUT1/6 and SREBP2 expression levels was also evaluated. The study elucidated the significant impact of GLUT1/6 on glioma cell functions, resulting in decreased glucose uptake. Moreover, it unveiled the regulatory role of SREBP2 in GLUT1 and GLUT6 transcription, alongside revealing differential expression of SREBP2 precursor and mature forms within gliomas. Following combined drug therapy, GLUT1/6 expression decreased, while the precursor form of SREBP2 increased, and mature SREBP2 reduced. This dual-drug treatment effectively modulated glioma cell energy metabolism. Subsequent in vivo experiments affirmed the augmented anti-tumor efficacy of combined drug therapy. Specifically, the synergistic action of metformin and simvastatin reshaped glioma metabolism, curbed malignant proliferation, promoted apoptosis, and demonstrated superior anti-tumor effects both in vitro and in vivo compared to individual administration of metformin or simvastatin. Importantly, the combination therapy achieved these effects at lower doses, rendering it a safer treatment option.

本研究旨在探讨二甲双胍和辛伐他汀联合疗法对胶质瘤恶性进展的抑制作用。研究特别探讨了作为转录因子的SREBP2的成熟如何影响胶质瘤细胞中GLUT1和GLUT6的表达。此外,研究还探讨了这种联合疗法对胶质瘤细胞的生物功能和能量代谢的影响。为了评估 GLUT1/6 的功能,研究人员使用了 sh-GLUT1/6 质粒。研究利用 CCK-8 试验确定了二甲双胍和辛伐他汀的半最大抑制浓度(IC50)。随后,研究人员使用明显低于IC50值的药物浓度,在体外和体内探讨了这些药物对胶质瘤代谢、增殖和凋亡的影响。研究还评估了药物治疗对 GLUT1/6 和 SREBP2 表达水平的影响。研究阐明了 GLUT1/6 对胶质瘤细胞功能的重大影响,导致葡萄糖摄取减少。此外,研究还揭示了 SREBP2 在 GLUT1 和 GLUT6 转录过程中的调控作用,同时揭示了 SREBP2 前体和成熟形式在胶质瘤中的不同表达。联合用药治疗后,GLUT1/6的表达减少,而SREBP2的前体形式增加,成熟的SREBP2减少。这种双重药物治疗有效地调节了胶质瘤细胞的能量代谢。随后的体内实验证实了联合用药的抗肿瘤效果。具体来说,与单独服用二甲双胍或辛伐他汀相比,二甲双胍和辛伐他汀的协同作用重塑了胶质瘤的新陈代谢,抑制了恶性增殖,促进了细胞凋亡,在体外和体内均显示出卓越的抗肿瘤效果。重要的是,联合疗法以较低的剂量就能达到这些效果,因此是一种更安全的治疗方案。
{"title":"Combined metformin and simvastatin therapy inhibits SREBP2 maturation and alters energy metabolism in glioma.","authors":"Xiaolong Qiao, Zixuan Wang, Yinan Chen, Nan Peng, Hongwei Zhang, Chaoshi Niu, Chuandong Cheng","doi":"10.1038/s41419-024-07169-5","DOIUrl":"10.1038/s41419-024-07169-5","url":null,"abstract":"<p><p>This study aims to explore the inhibitory effects of combined metformin and simvastatin therapy on the malignant progression of glioma. The research specifically examines how the maturation of SREBP2 as a transcription factor affects the expression of GLUT1 and GLUT6 in glioma cells. Additionally, it investigates the impact of this combination therapy on the biological functions and energy metabolism of glioma cells. To assess the functions of GLUT1/6, sh-GLUT1/6 plasmids were employed. The study determined the half-maximal inhibitory concentrations (IC50) of metformin and simvastatin using the CCK-8 assay. Subsequently, the effects of these drugs on glioma metabolism, proliferation, and apoptosis were explored in vitro and in vivo, using drug concentrations significantly lower than their respective IC50 values. The impact of drug treatment on GLUT1/6 and SREBP2 expression levels was also evaluated. The study elucidated the significant impact of GLUT1/6 on glioma cell functions, resulting in decreased glucose uptake. Moreover, it unveiled the regulatory role of SREBP2 in GLUT1 and GLUT6 transcription, alongside revealing differential expression of SREBP2 precursor and mature forms within gliomas. Following combined drug therapy, GLUT1/6 expression decreased, while the precursor form of SREBP2 increased, and mature SREBP2 reduced. This dual-drug treatment effectively modulated glioma cell energy metabolism. Subsequent in vivo experiments affirmed the augmented anti-tumor efficacy of combined drug therapy. Specifically, the synergistic action of metformin and simvastatin reshaped glioma metabolism, curbed malignant proliferation, promoted apoptosis, and demonstrated superior anti-tumor effects both in vitro and in vivo compared to individual administration of metformin or simvastatin. Importantly, the combination therapy achieved these effects at lower doses, rendering it a safer treatment option.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"809"},"PeriodicalIF":8.1,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550444/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PIGK defects induce apoptosis in Purkinje cells and acceleration of neuroectodermal differentiation. PIGK 缺陷会诱导浦肯野细胞凋亡并加速神经外胚层分化。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-09 DOI: 10.1038/s41419-024-07201-8
Siyi Chen, Jiali You, Xiaowei Zhou, Yan Li, Fang Liu, Yanling Teng, Hua Teng, Yunlong Li, Desheng Liang, Zhuo Li, Lingqian Wu

Biallelic mutations in PIGK cause GPI biosynthesis defect 22 (GPIBD22), characterized with developmental delay, hypotonia, and cerebellar atrophy. The understanding of the underlying causes is limited due to the lack of suitable disease models. To address this gap, we generated a mouse model with PIGK deficits, specifically in Purkinje cells (Pcp2-cko) and an induced pluripotent stem cell (iPSC) model using the c.87dupT mutant (KI) found in GPIBD22 patients. Pcp2-cko mice demonstrated cerebellar atrophy, ataxia and progressive Purkinje cells loss which were accompanied by increased apoptosis and neuroinflammation. Similarly, KI iPSCs exhibited increased apoptosis and accelerated neural rosette formation, indicating that PIGK defects could impact early neural differentiation that confirmed by the RNA-Seq results of neural progenitor cells (NPCs). The increased apoptosis and accelerated NPC differentiation in KI iPSCs are associated with excessive unfolded protein response (UPR) pathway activation, and can be rescued by UPR pathway inhibitor. Our study reveals potential pathogenic mechanism of GPIBD22 and providing new insights into the therapeutic strategy for GPIBD.

PIGK的双倍突变会导致GPI生物合成缺陷22(GPIBD22),表现为发育迟缓、肌张力低下和小脑萎缩。由于缺乏合适的疾病模型,人们对其根本原因的了解十分有限。为了填补这一空白,我们利用在 GPIBD22 患者中发现的 c.87dupT 突变体 (KI),建立了一个具有 PIGK 缺陷的小鼠模型,特别是在浦肯野细胞中(Pcp2-cko),以及一个诱导多能干细胞(iPSC)模型。Pcp2-cko小鼠表现出小脑萎缩、共济失调和进行性浦肯野细胞缺失,同时伴有细胞凋亡和神经炎症的增加。同样,KI iPSCs也表现出细胞凋亡增加和神经花环形成加速,这表明PIGK缺陷会影响早期神经分化,神经祖细胞(NPCs)的RNA-Seq结果证实了这一点。KI iPSCs细胞凋亡增加和NPC分化加速与过度激活未折叠蛋白反应(UPR)通路有关,UPR通路抑制剂可以挽救这种现象。我们的研究揭示了GPIBD22的潜在致病机制,并为GPIBD的治疗策略提供了新的见解。
{"title":"PIGK defects induce apoptosis in Purkinje cells and acceleration of neuroectodermal differentiation.","authors":"Siyi Chen, Jiali You, Xiaowei Zhou, Yan Li, Fang Liu, Yanling Teng, Hua Teng, Yunlong Li, Desheng Liang, Zhuo Li, Lingqian Wu","doi":"10.1038/s41419-024-07201-8","DOIUrl":"10.1038/s41419-024-07201-8","url":null,"abstract":"<p><p>Biallelic mutations in PIGK cause GPI biosynthesis defect 22 (GPIBD22), characterized with developmental delay, hypotonia, and cerebellar atrophy. The understanding of the underlying causes is limited due to the lack of suitable disease models. To address this gap, we generated a mouse model with PIGK deficits, specifically in Purkinje cells (Pcp2-cko) and an induced pluripotent stem cell (iPSC) model using the c.87dupT mutant (KI) found in GPIBD22 patients. Pcp2-cko mice demonstrated cerebellar atrophy, ataxia and progressive Purkinje cells loss which were accompanied by increased apoptosis and neuroinflammation. Similarly, KI iPSCs exhibited increased apoptosis and accelerated neural rosette formation, indicating that PIGK defects could impact early neural differentiation that confirmed by the RNA-Seq results of neural progenitor cells (NPCs). The increased apoptosis and accelerated NPC differentiation in KI iPSCs are associated with excessive unfolded protein response (UPR) pathway activation, and can be rescued by UPR pathway inhibitor. Our study reveals potential pathogenic mechanism of GPIBD22 and providing new insights into the therapeutic strategy for GPIBD.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"808"},"PeriodicalIF":8.1,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550446/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615633","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Inhibition of VDAC1 oligomerization blocks cysteine deprivation-induced ferroptosis via mitochondrial ROS suppression. 抑制 VDAC1 的寡聚化可通过抑制线粒体 ROS 阻断半胱氨酸剥夺诱导的铁中毒。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-09 DOI: 10.1038/s41419-024-07216-1
Se-Kyeong Jang, Se Hee Ahn, Gyeongmi Kim, Selim Kim, Jungil Hong, Ki Soo Park, In-Chul Park, Hyeon-Ok Jin

Ferroptosis, a regulated form of cell death dependent on reactive oxygen species (ROS), is characterized by iron accumulation and lethal lipid peroxidation. Mitochondria serve as the primary source of ROS and thus play a crucial role in ferroptosis initiation and execution. This study highlights the role of mitochondrial ROS and the significance of voltage-dependent anion channel 1 (VDAC1) oligomerization in ferroptosis induced by cysteine deprivation or ferroptosis-inducer RSL3. Our results demonstrate that the mitochondria-targeted antioxidants MitoQ and MitoT effectively block ferroptosis induction and that dysfunction of complex III of the mitochondrial electron transport chain contributes to ferroptosis induction. Pharmacological inhibitors that target VDAC1 oligomerization have emerged as potent suppressors of ferroptosis that reduce mitochondrial ROS production. These findings underscore the critical involvement of mitochondrial ROS production via complex III of the electron transport chain and the essential role of VDAC1 oligomerization in ferroptosis induced by cysteine deprivation or RSL3. This study deepens our understanding of the intricate molecular networks governing ferroptosis and provides insights into the development of novel therapeutic strategies targeting dysregulated cell death pathways.

铁中毒是一种依赖于活性氧(ROS)的细胞死亡调节形式,其特点是铁积累和致命的脂质过氧化。线粒体是 ROS 的主要来源,因此在铁变态反应的启动和执行过程中起着至关重要的作用。本研究强调了线粒体 ROS 的作用以及电压依赖性阴离子通道 1(VDAC1)寡聚化在半胱氨酸剥夺或铁变态反应诱导剂 RSL3 诱导的铁变态反应中的重要性。我们的研究结果表明,线粒体靶向抗氧化剂 MitoQ 和 MitoT 能有效阻断铁中毒诱导,线粒体电子传递链复合体 III 的功能障碍是铁中毒诱导的原因之一。以 VDAC1 寡聚化为靶点的药理抑制剂已成为减少线粒体 ROS 生成的铁变态反应的有效抑制剂。这些发现强调了线粒体通过电子传递链复合物 III 产生 ROS 的关键作用,以及 VDAC1 低聚化在半胱氨酸剥夺或 RSL3 诱导的铁变态反应中的重要作用。这项研究加深了我们对支配铁中毒的错综复杂的分子网络的理解,并为开发针对失调细胞死亡途径的新型治疗策略提供了见解。
{"title":"Inhibition of VDAC1 oligomerization blocks cysteine deprivation-induced ferroptosis via mitochondrial ROS suppression.","authors":"Se-Kyeong Jang, Se Hee Ahn, Gyeongmi Kim, Selim Kim, Jungil Hong, Ki Soo Park, In-Chul Park, Hyeon-Ok Jin","doi":"10.1038/s41419-024-07216-1","DOIUrl":"10.1038/s41419-024-07216-1","url":null,"abstract":"<p><p>Ferroptosis, a regulated form of cell death dependent on reactive oxygen species (ROS), is characterized by iron accumulation and lethal lipid peroxidation. Mitochondria serve as the primary source of ROS and thus play a crucial role in ferroptosis initiation and execution. This study highlights the role of mitochondrial ROS and the significance of voltage-dependent anion channel 1 (VDAC1) oligomerization in ferroptosis induced by cysteine deprivation or ferroptosis-inducer RSL3. Our results demonstrate that the mitochondria-targeted antioxidants MitoQ and MitoT effectively block ferroptosis induction and that dysfunction of complex III of the mitochondrial electron transport chain contributes to ferroptosis induction. Pharmacological inhibitors that target VDAC1 oligomerization have emerged as potent suppressors of ferroptosis that reduce mitochondrial ROS production. These findings underscore the critical involvement of mitochondrial ROS production via complex III of the electron transport chain and the essential role of VDAC1 oligomerization in ferroptosis induced by cysteine deprivation or RSL3. This study deepens our understanding of the intricate molecular networks governing ferroptosis and provides insights into the development of novel therapeutic strategies targeting dysregulated cell death pathways.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"811"},"PeriodicalIF":8.1,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550314/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615582","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Aurora B inhibitors promote RB hypophosphorylation and senescence independent of p53-dependent CDK2/4 inhibition. 极光 B 抑制剂可促进 RB 低磷酸化和衰老,而与 p53 依赖性 CDK2/4 抑制无关。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-11-09 DOI: 10.1038/s41419-024-07204-5
Shivam Vora, Ariel Andrew, Ramyashree Prasanna Kumar, Deborah Nazareth, Alexis Bonfim-Melo, Yoon Lim, Xin Yee Ong, Madushan Fernando, Yaowu He, John D Hooper, Nigel Aj McMillan, Jelena Urosevic, Jon Travers, Jamal Saeh, Sharad Kumar, Mathew Jk Jones, Brian Gabrielli

Aurora B kinase (AURKB) inhibitors have been trialled in a range of different tumour types but are not approved for any indication. Expression of the human papilloma virus (HPV) oncogenes and loss of retinoblastoma (RB) protein function has been reported to increase sensitivity to AURKB inhibitors but the mechanism of their contribution to sensitivity is poorly understood. Two commonly reported outcomes of AURKB inhibition are polyploidy and senescence, although their relationship is unclear. Here we have investigated the major cellular targets of the HPV E6 and E7, p53 and RB, to determine their contribution to AURKB inhibitor induced polyploidy and senescence. We demonstrate that polyploidy is a universal feature of AURKB inhibitor treatment in all cell types including normal primary cells, but the subsequent outcomes are controlled by RB and p53. We demonstrate that p53 by regulating p21 expression is required for an initial cell cycle arrest by inhibiting both CDK2 and CDK4 activity, but this arrest is only triggered after cells have undergone two failed mitosis and cytokinesis. However, cells can enter senescence in the absence of p53. RB is essential for AURKB inhibitor-induced senescence. AURKB inhibitor induces rapid hypophosphorylation of RB independent of inhibition of CDK2 or CDK4 kinases and p53. This work demonstrates that p53 activation determines the timing of senescence onset, but RB is indispensable for senescence.

极光 B 激酶(AURKB)抑制剂已在多种不同类型的肿瘤中试用,但尚未被批准用于任何适应症。据报道,人类乳头状瘤病毒(HPV)致癌基因的表达和视网膜母细胞瘤(RB)蛋白功能的丧失会增加对 AURKB 抑制剂的敏感性,但人们对它们增加敏感性的机制还不甚了解。通常报道的 AURKB 抑制的两种结果是多倍体和衰老,但它们之间的关系尚不清楚。在这里,我们研究了 HPV E6 和 E7 的主要细胞靶标 p53 和 RB,以确定它们对 AURKB 抑制剂诱导的多倍体和衰老的贡献。我们证明,多倍体是 AURKB 抑制剂处理包括正常原代细胞在内的所有细胞类型的普遍特征,但随后的结果受 RB 和 p53 控制。我们证明,p53 通过调节 p21 的表达,抑制 CDK2 和 CDK4 的活性,从而实现初始细胞周期的停滞,但这种停滞只有在细胞经历了两次失败的有丝分裂和细胞分裂后才会触发。然而,在没有 p53 的情况下,细胞也会进入衰老期。RB对AURKB抑制剂诱导的衰老至关重要。AURKB 抑制剂可诱导 RB 快速低磷酸化,而与 CDK2 或 CDK4 激酶和 p53 的抑制作用无关。这项工作表明,p53的激活决定了衰老开始的时间,但RB对衰老是不可或缺的。
{"title":"Aurora B inhibitors promote RB hypophosphorylation and senescence independent of p53-dependent CDK2/4 inhibition.","authors":"Shivam Vora, Ariel Andrew, Ramyashree Prasanna Kumar, Deborah Nazareth, Alexis Bonfim-Melo, Yoon Lim, Xin Yee Ong, Madushan Fernando, Yaowu He, John D Hooper, Nigel Aj McMillan, Jelena Urosevic, Jon Travers, Jamal Saeh, Sharad Kumar, Mathew Jk Jones, Brian Gabrielli","doi":"10.1038/s41419-024-07204-5","DOIUrl":"10.1038/s41419-024-07204-5","url":null,"abstract":"<p><p>Aurora B kinase (AURKB) inhibitors have been trialled in a range of different tumour types but are not approved for any indication. Expression of the human papilloma virus (HPV) oncogenes and loss of retinoblastoma (RB) protein function has been reported to increase sensitivity to AURKB inhibitors but the mechanism of their contribution to sensitivity is poorly understood. Two commonly reported outcomes of AURKB inhibition are polyploidy and senescence, although their relationship is unclear. Here we have investigated the major cellular targets of the HPV E6 and E7, p53 and RB, to determine their contribution to AURKB inhibitor induced polyploidy and senescence. We demonstrate that polyploidy is a universal feature of AURKB inhibitor treatment in all cell types including normal primary cells, but the subsequent outcomes are controlled by RB and p53. We demonstrate that p53 by regulating p21 expression is required for an initial cell cycle arrest by inhibiting both CDK2 and CDK4 activity, but this arrest is only triggered after cells have undergone two failed mitosis and cytokinesis. However, cells can enter senescence in the absence of p53. RB is essential for AURKB inhibitor-induced senescence. AURKB inhibitor induces rapid hypophosphorylation of RB independent of inhibition of CDK2 or CDK4 kinases and p53. This work demonstrates that p53 activation determines the timing of senescence onset, but RB is indispensable for senescence.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":"15 11","pages":"810"},"PeriodicalIF":8.1,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11550316/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615512","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
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:481959085
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