首页 > 最新文献

Cell Death and Differentiation最新文献

英文 中文
AMDHD1 acts as a tumor suppressor and contributes to activation of TGF-β signaling pathway in cholangiocarcinoma. AMDHD1 是一种肿瘤抑制因子,有助于激活胆管癌中的 TGF-β 信号通路。
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-14 DOI: 10.1038/s41418-024-01361-y
Zuyi Ma, Jia Sun, Zhenchong Li, Shanzhou Huang, Binglu Li

Cholangiocarcinoma (CCA) is a malignant tumor of the digestive system, characterized by its aggressive behavior and the absence of effective therapeutic biomarkers. Although recent studies have implicated AMDHD1 in tumor formation, its role in CCA development has been insufficiently explored. We utilized multiple bioinformatic datasets alongside 108 clinical samples to examine AMDHD1 expression in CCA. Then, in vitro and in vivo experiments were conducted to assess its impact on tumor growth and metastasis. Furthermore, proteomic analysis and immunoprecipitation mass spectrometry were employed to identify the downstream effectors of AMDHD1. We discovered that AMDHD1 was down-regulated in CCA and this down-regulation was associated with adverse clinicopathological features and prognosis. We also demonstrated that overexpression of AMDHD1 hindered G1/S progression in the cell cycle and promoted apoptosis, thereby inhibiting tumor growth and metastasis. Mechanistically, we found that AMDHD1 operated in a TGF-β-dependent manner and the inhibition of TGF-β signaling abrogated the effect of AMDHD1 overexpression on CCA cells. Specifically, AMDHD1 inhibited the ubiquitination and degradation of the SMAD4 protein through binding to the MH2 domain and synergistically enhanced SMAD2/3 phosphorylation, which activated of TGF-β signaling pathway and resulted in the suppression of CCA cell proliferation and migration. Our study identifies AMDHD1 as a significant prognostic biomarker and a tumor suppressor in CCA. It underscores the pivotal role of the AMDHD1/TGF-β signaling pathway in the development and progression of CCA.

胆管癌(CCA)是消化系统的一种恶性肿瘤,其特点是具有侵袭性和缺乏有效的治疗生物标志物。尽管最近的研究表明 AMDHD1 与肿瘤的形成有关,但其在 CCA 发展过程中的作用尚未得到充分探讨。我们利用多个生物信息数据集和 108 个临床样本研究了 AMDHD1 在 CCA 中的表达。然后,我们进行了体外和体内实验,评估其对肿瘤生长和转移的影响。此外,我们还采用了蛋白质组分析和免疫沉淀质谱法来确定AMDHD1的下游效应因子。我们发现 AMDHD1 在 CCA 中被下调,而这种下调与不良的临床病理特征和预后有关。我们还证实,AMDHD1的过表达会阻碍细胞周期的G1/S进展,促进细胞凋亡,从而抑制肿瘤的生长和转移。从机理上讲,我们发现AMDHD1以依赖于TGF-β的方式运行,而抑制TGF-β信号传导可减弱AMDHD1过表达对CCA细胞的影响。具体而言,AMDHD1通过与MH2结构域结合抑制SMAD4蛋白的泛素化和降解,并协同增强SMAD2/3磷酸化,从而激活TGF-β信号通路,抑制CCA细胞的增殖和迁移。我们的研究发现,AMDHD1是CCA的重要预后生物标志物和肿瘤抑制因子。它强调了AMDHD1/TGF-β信号通路在CCA的发生和发展中的关键作用。
{"title":"AMDHD1 acts as a tumor suppressor and contributes to activation of TGF-β signaling pathway in cholangiocarcinoma.","authors":"Zuyi Ma, Jia Sun, Zhenchong Li, Shanzhou Huang, Binglu Li","doi":"10.1038/s41418-024-01361-y","DOIUrl":"https://doi.org/10.1038/s41418-024-01361-y","url":null,"abstract":"<p><p>Cholangiocarcinoma (CCA) is a malignant tumor of the digestive system, characterized by its aggressive behavior and the absence of effective therapeutic biomarkers. Although recent studies have implicated AMDHD1 in tumor formation, its role in CCA development has been insufficiently explored. We utilized multiple bioinformatic datasets alongside 108 clinical samples to examine AMDHD1 expression in CCA. Then, in vitro and in vivo experiments were conducted to assess its impact on tumor growth and metastasis. Furthermore, proteomic analysis and immunoprecipitation mass spectrometry were employed to identify the downstream effectors of AMDHD1. We discovered that AMDHD1 was down-regulated in CCA and this down-regulation was associated with adverse clinicopathological features and prognosis. We also demonstrated that overexpression of AMDHD1 hindered G1/S progression in the cell cycle and promoted apoptosis, thereby inhibiting tumor growth and metastasis. Mechanistically, we found that AMDHD1 operated in a TGF-β-dependent manner and the inhibition of TGF-β signaling abrogated the effect of AMDHD1 overexpression on CCA cells. Specifically, AMDHD1 inhibited the ubiquitination and degradation of the SMAD4 protein through binding to the MH2 domain and synergistically enhanced SMAD2/3 phosphorylation, which activated of TGF-β signaling pathway and resulted in the suppression of CCA cell proliferation and migration. Our study identifies AMDHD1 as a significant prognostic biomarker and a tumor suppressor in CCA. It underscores the pivotal role of the AMDHD1/TGF-β signaling pathway in the development and progression of CCA.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":13.7,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141981814","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
LIM domain only 7: a novel driver of immune evasion through regulatory T cell differentiation and chemotaxis in pancreatic ductal adenocarcinoma. 仅有 LIM 结构域的 7:胰腺导管腺癌中通过调节性 T 细胞分化和趋化作用逃避免疫的新型驱动因素。
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-14 DOI: 10.1038/s41418-024-01358-7
Shangnan Dai, Yunpeng Peng, Guangfu Wang, Chongfa Chen, Qiuyang Chen, Lingdi Yin, Han Yan, Kai Zhang, Min Tu, Zipeng Lu, Jishu Wei, Qiang Li, Junli Wu, Kuirong Jiang, Yi Zhu, Yi Miao

With advancements in genomics and immunology, immunotherapy has emerged as a revolutionary strategy for tumor treatment. However, pancreatic ductal adenocarcinoma (PDAC), an immunologically "cold" tumor, exhibits limited responsiveness to immunotherapy. This study aimed to address the urgent need to uncover PDAC's immune microenvironment heterogeneity and identify the molecular mechanisms driving immune evasion. Using single-cell RNA sequencing datasets and spatial proteomics, we discovered LIM domain only 7 (LMO7) in PDAC cells as a previously unrecognized driver of immune evasion through Treg cell enrichment. LMO7 was positively correlated with infiltrating regulatory T cells (Tregs) and dysfunctional CD8+ T cells. A series of in vitro and in vivo experiments demonstrated LMO7's significant role in promoting Treg cell differentiation and chemotaxis while inhibiting CD8+ T cells and natural killer cell cytotoxicity. Mechanistically, LMO7, through its LIM domain, directly bound and promoted the ubiquitination and degradation of Foxp1. Foxp1 negatively regulated transforming growth factor-beta (TGF-β) and C-C motif chemokine ligand 5 (CCL5) expression by binding to sites 2 and I/III, respectively. Elevated TGF-β and CCL5 levels contribute to Treg cell enrichment, inducing immune evasion in PDAC. Combined treatment with TGF-β/CCL5 antibodies, along with LMO7 inhibition, effectively reversed immune evasion in PDAC, activated the immune response, and prolonged mouse survival. Therefore, this study identified LMO7 as a novel facilitator in driving immune evasion by promoting Treg cell enrichment and inhibiting cytotoxic effector functions. Targeting the LMO7-Foxp1-TGF-β/CCL5 axis holds promise as a therapeutic strategy for PDAC. Graphical abstract revealing LMO7 as a novel facilitator in driving immune evasion by promoting Tregs differentiation and chemotaxis, inducing CD8+ T/natural killer cells inhibition.

随着基因组学和免疫学的进步,免疫疗法已成为一种革命性的肿瘤治疗策略。然而,胰腺导管腺癌(PDAC)是一种免疫 "冷 "肿瘤,对免疫疗法的反应有限。本研究旨在满足揭示PDAC免疫微环境异质性和鉴定驱动免疫逃避的分子机制的迫切需要。利用单细胞RNA测序数据集和空间蛋白质组学,我们在PDAC细胞中发现了LIM domain only 7 (LMO7),它是之前未被发现的通过Treg细胞富集驱动免疫逃避的因素。LMO7 与浸润性调节性 T 细胞(Tregs)和功能失调的 CD8+ T 细胞呈正相关。一系列体外和体内实验证明,LMO7 在促进 Treg 细胞分化和趋化,同时抑制 CD8+ T 细胞和自然杀伤细胞细胞毒性方面发挥着重要作用。从机理上讲,LMO7 通过其 LIM 结构域直接结合并促进 Foxp1 的泛素化和降解。Foxp1 通过分别与位点 2 和位点 I/III 结合,负向调节转化生长因子-β(TGF-β)和 C-C motif 趋化因子配体 5(CCL5)的表达。TGF-β和CCL5水平的升高有助于Treg细胞的富集,诱导PDAC的免疫逃避。TGF-β/CCL5抗体与LMO7抑制剂联合治疗可有效逆转PDAC的免疫逃避,激活免疫反应,延长小鼠存活时间。因此,本研究发现LMO7是通过促进Treg细胞富集和抑制细胞毒性效应因子功能来驱动免疫逃避的新型促进因子。以LMO7-Foxp1-TGF-β/CCL5轴为靶点有望成为PDAC的治疗策略。图解摘要揭示了 LMO7 通过促进集落细胞分化和趋化,诱导 CD8+ T/自然杀伤细胞抑制,成为驱动免疫逃避的新型促进因子。
{"title":"LIM domain only 7: a novel driver of immune evasion through regulatory T cell differentiation and chemotaxis in pancreatic ductal adenocarcinoma.","authors":"Shangnan Dai, Yunpeng Peng, Guangfu Wang, Chongfa Chen, Qiuyang Chen, Lingdi Yin, Han Yan, Kai Zhang, Min Tu, Zipeng Lu, Jishu Wei, Qiang Li, Junli Wu, Kuirong Jiang, Yi Zhu, Yi Miao","doi":"10.1038/s41418-024-01358-7","DOIUrl":"https://doi.org/10.1038/s41418-024-01358-7","url":null,"abstract":"<p><p>With advancements in genomics and immunology, immunotherapy has emerged as a revolutionary strategy for tumor treatment. However, pancreatic ductal adenocarcinoma (PDAC), an immunologically \"cold\" tumor, exhibits limited responsiveness to immunotherapy. This study aimed to address the urgent need to uncover PDAC's immune microenvironment heterogeneity and identify the molecular mechanisms driving immune evasion. Using single-cell RNA sequencing datasets and spatial proteomics, we discovered LIM domain only 7 (LMO7) in PDAC cells as a previously unrecognized driver of immune evasion through Treg cell enrichment. LMO7 was positively correlated with infiltrating regulatory T cells (Tregs) and dysfunctional CD8<sup>+</sup> T cells. A series of in vitro and in vivo experiments demonstrated LMO7's significant role in promoting Treg cell differentiation and chemotaxis while inhibiting CD8<sup>+</sup> T cells and natural killer cell cytotoxicity. Mechanistically, LMO7, through its LIM domain, directly bound and promoted the ubiquitination and degradation of Foxp1. Foxp1 negatively regulated transforming growth factor-beta (TGF-β) and C-C motif chemokine ligand 5 (CCL5) expression by binding to sites 2 and I/III, respectively. Elevated TGF-β and CCL5 levels contribute to Treg cell enrichment, inducing immune evasion in PDAC. Combined treatment with TGF-β/CCL5 antibodies, along with LMO7 inhibition, effectively reversed immune evasion in PDAC, activated the immune response, and prolonged mouse survival. Therefore, this study identified LMO7 as a novel facilitator in driving immune evasion by promoting Treg cell enrichment and inhibiting cytotoxic effector functions. Targeting the LMO7-Foxp1-TGF-β/CCL5 axis holds promise as a therapeutic strategy for PDAC. Graphical abstract revealing LMO7 as a novel facilitator in driving immune evasion by promoting Tregs differentiation and chemotaxis, inducing CD8<sup>+</sup> T/natural killer cells inhibition.</p>","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":" ","pages":""},"PeriodicalIF":13.7,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141981815","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
Rewiring cancer cell death: LPCAT1 shapes lipid composition and ferroptosis resistance 重构癌细胞死亡:LPCAT1 塑造脂质组成和铁中毒抗性
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-13 DOI: 10.1038/s41418-024-01364-9
Hyemin Lee, Li Zhuang, Boyi Gan
{"title":"Rewiring cancer cell death: LPCAT1 shapes lipid composition and ferroptosis resistance","authors":"Hyemin Lee,&nbsp;Li Zhuang,&nbsp;Boyi Gan","doi":"10.1038/s41418-024-01364-9","DOIUrl":"10.1038/s41418-024-01364-9","url":null,"abstract":"","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 9","pages":"1101-1103"},"PeriodicalIF":13.7,"publicationDate":"2024-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141973804","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The PRMT6/STAT1/ACSL1 axis promotes ferroptosis in diabetic nephropathy PRMT6/STAT1/ACSL1 轴促进糖尿病肾病中的铁蛋白沉积。
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-13 DOI: 10.1038/s41418-024-01357-8
Jia Hong, Xue Li, Yingxiang Hao, Hongjiao Xu, Lang Yu, Zhipeng Meng, Jianhai Zhang, Minmin Zhu
Hyperglycaemia-induced ferroptosis is a significant contributor to kidney dysfunction in diabetic nephropathy (DN) patients. In addition, targeting ferroptosis has clinical implications for the treatment of DN. However, effective therapeutic targets for ferroptosis have not been identified. In this study, we aimed to explore the precise role of protein arginine methyltransferase 6 (PRMT6) in regulating ferroptosis in DN. In the present study, we utilized a mouse DN model consisting of both wild-type and PRMT6-knockout (PRMT6−/−) mice. Transcriptomic and lipidomic analyses, along with various molecular biological methodologies, were used to determine the potential mechanism by which PRMT6 regulates ferroptosis in DN. Our results indicate that PRMT6 downregulation participates in kidney dysfunction and renal cell death via the modulation of ferroptosis in DN. Moreover, PRMT6 reduction induced lipid peroxidation by upregulating acyl-CoA synthetase long-chain family member 1 (ACSL1) expression, ultimately contributing to ferroptosis. Furthermore, we investigated the molecular mechanism by which PRMT6 interacts with signal transducer and activator of transcription 1 (STAT1) to jointly regulate ACSL1 transcription. Additionally, treatment with the STAT1-specific inhibitor fludarabine delayed DN progression. Furthermore, we observed that PRMT6 and STAT1 synergistically regulate ACSL1 transcription to mediate ferroptosis in hyperglycaemic cells. Our study demonstrated that PRMT6 and STAT1 comodulate ACSL1 transcription to induce the production of phospholipid-polyunsaturated fatty acids (PL-PUFAs), thus participating in ferroptosis in DN. These findings suggest that the PRMT6/STAT1/ACSL1 axis is a new therapeutic target for the prevention and treatment of DN.
高血糖诱导的高铁血症是导致糖尿病肾病(DN)患者肾功能障碍的重要原因。此外,针对高铁血症的治疗对治疗糖尿病肾病也有临床意义。然而,目前尚未发现针对铁蛋白沉积的有效治疗靶点。在本研究中,我们旨在探索蛋白精氨酸甲基转移酶 6(PRMT6)在 DN 中调控铁凋亡的确切作用。在本研究中,我们使用了由野生型和 PRMT6 基因敲除(PRMT6-/-)小鼠组成的小鼠 DN 模型。通过转录组和脂质组分析以及各种分子生物学方法,我们确定了 PRMT6 在 DN 中调控铁突变的潜在机制。我们的研究结果表明,PRMT6 的下调通过调节 DN 中的铁蛋白沉积参与了肾功能障碍和肾细胞死亡。此外,PRMT6 下调通过上调酰基-CoA 合成酶长链家族成员 1(ACSL1)的表达诱导脂质过氧化,最终导致铁变态反应。此外,我们还研究了 PRMT6 与信号转导子和转录激活子 1(STAT1)相互作用共同调控 ACSL1 转录的分子机制。此外,用 STAT1 特异性抑制剂氟达拉滨治疗可延缓 DN 的进展。此外,我们还观察到 PRMT6 和 STAT1 能协同调控 ACSL1 的转录,从而介导高血糖细胞的铁变态反应。我们的研究表明,PRMT6和STAT1共同调控ACSL1转录,诱导磷脂-多不饱和脂肪酸(PL-PUFAs)的产生,从而参与了DN中的铁突变。这些研究结果表明,PRMT6/STAT1/ACSL1 轴是预防和治疗 DN 的一个新的治疗靶点。
{"title":"The PRMT6/STAT1/ACSL1 axis promotes ferroptosis in diabetic nephropathy","authors":"Jia Hong,&nbsp;Xue Li,&nbsp;Yingxiang Hao,&nbsp;Hongjiao Xu,&nbsp;Lang Yu,&nbsp;Zhipeng Meng,&nbsp;Jianhai Zhang,&nbsp;Minmin Zhu","doi":"10.1038/s41418-024-01357-8","DOIUrl":"10.1038/s41418-024-01357-8","url":null,"abstract":"Hyperglycaemia-induced ferroptosis is a significant contributor to kidney dysfunction in diabetic nephropathy (DN) patients. In addition, targeting ferroptosis has clinical implications for the treatment of DN. However, effective therapeutic targets for ferroptosis have not been identified. In this study, we aimed to explore the precise role of protein arginine methyltransferase 6 (PRMT6) in regulating ferroptosis in DN. In the present study, we utilized a mouse DN model consisting of both wild-type and PRMT6-knockout (PRMT6−/−) mice. Transcriptomic and lipidomic analyses, along with various molecular biological methodologies, were used to determine the potential mechanism by which PRMT6 regulates ferroptosis in DN. Our results indicate that PRMT6 downregulation participates in kidney dysfunction and renal cell death via the modulation of ferroptosis in DN. Moreover, PRMT6 reduction induced lipid peroxidation by upregulating acyl-CoA synthetase long-chain family member 1 (ACSL1) expression, ultimately contributing to ferroptosis. Furthermore, we investigated the molecular mechanism by which PRMT6 interacts with signal transducer and activator of transcription 1 (STAT1) to jointly regulate ACSL1 transcription. Additionally, treatment with the STAT1-specific inhibitor fludarabine delayed DN progression. Furthermore, we observed that PRMT6 and STAT1 synergistically regulate ACSL1 transcription to mediate ferroptosis in hyperglycaemic cells. Our study demonstrated that PRMT6 and STAT1 comodulate ACSL1 transcription to induce the production of phospholipid-polyunsaturated fatty acids (PL-PUFAs), thus participating in ferroptosis in DN. These findings suggest that the PRMT6/STAT1/ACSL1 axis is a new therapeutic target for the prevention and treatment of DN.","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 11","pages":"1561-1575"},"PeriodicalIF":13.7,"publicationDate":"2024-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141970698","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
Vps34 sustains Treg cell survival and function via regulating intracellular redox homeostasis Vps34 通过调节细胞内氧化还原平衡维持 Treg 细胞的存活和功能
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41418-024-01353-y
Peiran Feng, Quanli Yang, Liang Luo, Zerong Guan, Jiamin Fu, Mingyue Zhao, Wanqing Meng, Shuo Wan, Junming He, Zhizhong Li, Guang Wang, Guodong Sun, Zhongjun Dong, Meixiang Yang
The survival and suppressive function of regulatory T (Treg) cells rely on various intracellular metabolic and physiological processes. Our study demonstrates that Vps34 plays a critical role in maintaining Treg cell homeostasis and function by regulating cellular metabolic activities. Disruption of Vps34 in Treg cells leads to spontaneous fatal systemic autoimmune disorder and multi-tissue inflammatory damage, accompanied by a reduction in the number of Treg cells, particularly eTreg cells with highly immunosuppressive activity. Mechanistically, the poor survival of Vps34-deficient Treg cells is attributed to impaired endocytosis, intracellular vesicular trafficking and autophagosome formation, which further results in enhanced mitochondrial respiration and excessive ROS production. Removal of excessive ROS can effectively rescue the death of Vps34-deficient Treg cells. Functionally, acute deletion of Vps34 within established Treg cells enhances anti-tumor immunity in a malignant melanoma model by boosting T-cell-mediated anti-tumor activity. Overall, our results underscore the pivotal role played by Vps34 in orchestrating Treg cell homeostasis and function towards establishing immune homeostasis and tolerance.
调节性 T(Treg)细胞的存活和抑制功能依赖于各种细胞内代谢和生理过程。我们的研究表明,Vps34 通过调节细胞代谢活动,在维持 Treg 细胞稳态和功能方面发挥着关键作用。Treg细胞中的Vps34被破坏会导致自发性致命的系统性自身免疫紊乱和多组织炎症损伤,同时伴随着Treg细胞数量的减少,尤其是具有高度免疫抑制活性的eTreg细胞。从机理上讲,Vps34缺陷的Treg细胞存活率低是由于内吞、细胞内囊泡运输和自噬体形成受损,从而进一步导致线粒体呼吸增强和ROS产生过多。清除过量的 ROS 可有效挽救 Vps34 缺陷 Treg 细胞的死亡。从功能上讲,在恶性黑色素瘤模型中,在已建立的 Treg 细胞中急性缺失 Vps34 可增强 T 细胞介导的抗肿瘤活性,从而增强抗肿瘤免疫力。总之,我们的研究结果凸显了 Vps34 在协调 Treg 细胞稳态和功能以建立免疫稳态和耐受性方面发挥的关键作用。
{"title":"Vps34 sustains Treg cell survival and function via regulating intracellular redox homeostasis","authors":"Peiran Feng,&nbsp;Quanli Yang,&nbsp;Liang Luo,&nbsp;Zerong Guan,&nbsp;Jiamin Fu,&nbsp;Mingyue Zhao,&nbsp;Wanqing Meng,&nbsp;Shuo Wan,&nbsp;Junming He,&nbsp;Zhizhong Li,&nbsp;Guang Wang,&nbsp;Guodong Sun,&nbsp;Zhongjun Dong,&nbsp;Meixiang Yang","doi":"10.1038/s41418-024-01353-y","DOIUrl":"10.1038/s41418-024-01353-y","url":null,"abstract":"The survival and suppressive function of regulatory T (Treg) cells rely on various intracellular metabolic and physiological processes. Our study demonstrates that Vps34 plays a critical role in maintaining Treg cell homeostasis and function by regulating cellular metabolic activities. Disruption of Vps34 in Treg cells leads to spontaneous fatal systemic autoimmune disorder and multi-tissue inflammatory damage, accompanied by a reduction in the number of Treg cells, particularly eTreg cells with highly immunosuppressive activity. Mechanistically, the poor survival of Vps34-deficient Treg cells is attributed to impaired endocytosis, intracellular vesicular trafficking and autophagosome formation, which further results in enhanced mitochondrial respiration and excessive ROS production. Removal of excessive ROS can effectively rescue the death of Vps34-deficient Treg cells. Functionally, acute deletion of Vps34 within established Treg cells enhances anti-tumor immunity in a malignant melanoma model by boosting T-cell-mediated anti-tumor activity. Overall, our results underscore the pivotal role played by Vps34 in orchestrating Treg cell homeostasis and function towards establishing immune homeostasis and tolerance.","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 11","pages":"1519-1533"},"PeriodicalIF":13.7,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141904492","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
Novel role for Ddx39 in differentiation and telomere length regulation of embryonic stem cells Ddx39 在胚胎干细胞分化和端粒长度调节中的新作用
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-06 DOI: 10.1038/s41418-024-01354-x
Shanshan Nai, Meijie Wang, Jiao Yang, Bin Ling, Qiman Dong, Xiaoqiong Yang, Xiaoling Du, Man Lu, Lin Liu, Zhongbo Yu, Lingyi Chen
Erk signaling is indispensable for the self-renewal and differentiation of mouse embryonic stem cells (ESCs), as well as telomere homeostasis. But how Erk regulates these biological processes remains unclear. We identified 132 Erk2 interacting proteins by co-immunoprecipitation and mass spectrometric analysis, and focused on Ddx39 as a potential Erk2 substrate. We demonstrated that Erk2 phosphorylates Ddx39 on Y132 and Y138. Ddx39 knockout (KO) ESCs are defective in differentiation, due to reduced H3K27ac level upon differentiation. Phosphorylation of Ddx39 promotes the recruitment of Hat1 to acetylate H3K27 and activate differentiation genes. In addition, Ddx39 KO leads to telomere elongation in ESCs. Ddx39 is recruited to telomeres by the telomere-binding protein Trf1, consequently disrupting the DNA loop formed by Trf1 and suppressing the alternative lengthening of telomeres (ALT). Phosphorylation of Ddx39 weakens its interaction with Trf1, releasing it from telomeres. Thus, ALT activity is enhanced, and telomeres are elongated. Altogether, our studies reveal an essential role of Ddx39 in the differentiation and telomere homeostasis of ESCs.
Erk信号对于小鼠胚胎干细胞(ESC)的自我更新和分化以及端粒稳态是不可或缺的。但Erk如何调控这些生物学过程仍不清楚。我们通过共免疫沉淀和质谱分析鉴定了132个Erk2相互作用蛋白,并重点研究了作为Erk2潜在底物的Ddx39。我们证明了Erk2能使Ddx39在Y132和Y138上磷酸化。Ddx39基因敲除(KO)的ESC在分化过程中会出现H3K27ac水平降低,从而导致分化缺陷。Ddx39的磷酸化会促进Hat1的招募,使H3K27乙酰化并激活分化基因。此外,Ddx39 KO会导致ESC端粒延长。Ddx39被端粒结合蛋白Trf1招募到端粒上,从而破坏了Trf1形成的DNA环,抑制了端粒的替代性延长(ALT)。Ddx39 的磷酸化削弱了它与 Trf1 的相互作用,使其从端粒中释放出来。因此,ALT 活性增强,端粒延长。总之,我们的研究揭示了 Ddx39 在造血干细胞的分化和端粒稳态中的重要作用。
{"title":"Novel role for Ddx39 in differentiation and telomere length regulation of embryonic stem cells","authors":"Shanshan Nai,&nbsp;Meijie Wang,&nbsp;Jiao Yang,&nbsp;Bin Ling,&nbsp;Qiman Dong,&nbsp;Xiaoqiong Yang,&nbsp;Xiaoling Du,&nbsp;Man Lu,&nbsp;Lin Liu,&nbsp;Zhongbo Yu,&nbsp;Lingyi Chen","doi":"10.1038/s41418-024-01354-x","DOIUrl":"10.1038/s41418-024-01354-x","url":null,"abstract":"Erk signaling is indispensable for the self-renewal and differentiation of mouse embryonic stem cells (ESCs), as well as telomere homeostasis. But how Erk regulates these biological processes remains unclear. We identified 132 Erk2 interacting proteins by co-immunoprecipitation and mass spectrometric analysis, and focused on Ddx39 as a potential Erk2 substrate. We demonstrated that Erk2 phosphorylates Ddx39 on Y132 and Y138. Ddx39 knockout (KO) ESCs are defective in differentiation, due to reduced H3K27ac level upon differentiation. Phosphorylation of Ddx39 promotes the recruitment of Hat1 to acetylate H3K27 and activate differentiation genes. In addition, Ddx39 KO leads to telomere elongation in ESCs. Ddx39 is recruited to telomeres by the telomere-binding protein Trf1, consequently disrupting the DNA loop formed by Trf1 and suppressing the alternative lengthening of telomeres (ALT). Phosphorylation of Ddx39 weakens its interaction with Trf1, releasing it from telomeres. Thus, ALT activity is enhanced, and telomeres are elongated. Altogether, our studies reveal an essential role of Ddx39 in the differentiation and telomere homeostasis of ESCs.","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 11","pages":"1534-1544"},"PeriodicalIF":13.7,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896902","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
Phospholipid peroxidation in macrophage confers tumor resistance by suppressing phagocytic capability towards ferroptotic cells 巨噬细胞中的磷脂过氧化反应可抑制对铁锈细胞的吞噬能力,从而产生抗肿瘤能力。
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-05 DOI: 10.1038/s41418-024-01351-0
Xiang Luo, Hai-Biao Gong, Zi-Chun Li, Dong-Dong Li, Zi-Xuan Li, Jie Sun, Chang-Yu Yan, Rui-Ting Huang, Yue Feng, Shu-Rui Chen, Yun-Feng Cao, Mingxian Liu, Rong Wang, Feng Huang, Wan-Yang Sun, Hiroshi Kurihara, Wen-Jun Duan, Lei Liang, Wen Jin, Yan-Ping Wu, Rong-Rong He, Yi-Fang Li
Ferroptosis holds significant potential for application in cancer therapy. However, ferroptosis inducers are not cell-specific and can cause phospholipid peroxidation in both tumor and non-tumor cells. This limitation greatly restricts the use of ferroptosis therapy as a safe and effective anticancer strategy. Our previous study demonstrated that macrophages can engulf ferroptotic cells through Toll-like receptor 2 (TLR2). Despite this advancement, the precise mechanism by which phospholipid peroxidation in macrophages affects their phagocytotic capability during treatment of tumors with ferroptotic agents is still unknown. Here, we utilized flow sorting combined with redox phospholipidomics to determine that phospholipid peroxidation in tumor microenvironment (TME) macrophages impaired the macrophages ability to eliminate ferroptotic tumor cells by phagocytosis, ultimately fostering tumor resistance to ferroptosis therapy. Mechanistically, the accumulation of phospholipid peroxidation in the macrophage endoplasmic reticulum (ER) repressed TLR2 trafficking to the plasma membrane and caused its retention in the ER by disrupting the interaction between TLR2 and its chaperone CNPY3. Subsequently, this ER-retained TLR2 recruited E3 ligase MARCH6 and initiated the proteasome-dependent degradation. Using redox phospholipidomics, we identified 1-steaoryl-2-15-HpETE-sn-glycero-3-phosphatidylethanolamine (SAPE-OOH) as the crucial mediator of these effects. Conclusively, our discovery elucidates a novel molecular mechanism underlying macrophage phospholipid peroxidation-induced tumor resistance to ferroptosis therapy and highlights the TLR2-MARCH6 axis as a potential therapeutic target for cancer therapy.
铁氧化在癌症治疗中具有巨大的应用潜力。然而,铁突变诱导剂并非细胞特异性诱导剂,可导致肿瘤细胞和非肿瘤细胞的磷脂过氧化。这一局限性极大地限制了铁突变疗法作为一种安全有效的抗癌策略的应用。我们之前的研究表明,巨噬细胞可以通过 Toll 样受体 2(TLR2)吞噬铁氧体细胞。尽管取得了这一进展,但巨噬细胞中的磷脂过氧化影响其在使用铁氧体药物治疗肿瘤过程中的吞噬能力的确切机制仍不清楚。在这里,我们利用流式细胞分拣技术结合氧化还原磷脂组学确定了肿瘤微环境(TME)巨噬细胞中的磷脂过氧化反应会损害巨噬细胞通过吞噬作用消灭嗜铁肿瘤细胞的能力,最终导致肿瘤对嗜铁疗法产生耐药性。从机理上讲,巨噬细胞内质网(ER)中磷脂过氧化物的积累抑制了 TLR2 向质膜的迁移,并通过破坏 TLR2 与其伴侣 CNPY3 之间的相互作用使其滞留在 ER 中。随后,保留在ER中的TLR2招募E3连接酶MARCH6,并启动蛋白酶体依赖性降解。利用氧化还原磷脂组学,我们发现 1-steaoryl-2-15-HpETE-sn-glycero-3-phosphatidylethanolamine(SAPE-OOH)是这些效应的关键介质。最后,我们的发现阐明了巨噬细胞磷脂过氧化诱导的肿瘤抗铁治疗的新分子机制,并强调了TLR2-MARCH6轴是癌症治疗的潜在治疗靶点。
{"title":"Phospholipid peroxidation in macrophage confers tumor resistance by suppressing phagocytic capability towards ferroptotic cells","authors":"Xiang Luo,&nbsp;Hai-Biao Gong,&nbsp;Zi-Chun Li,&nbsp;Dong-Dong Li,&nbsp;Zi-Xuan Li,&nbsp;Jie Sun,&nbsp;Chang-Yu Yan,&nbsp;Rui-Ting Huang,&nbsp;Yue Feng,&nbsp;Shu-Rui Chen,&nbsp;Yun-Feng Cao,&nbsp;Mingxian Liu,&nbsp;Rong Wang,&nbsp;Feng Huang,&nbsp;Wan-Yang Sun,&nbsp;Hiroshi Kurihara,&nbsp;Wen-Jun Duan,&nbsp;Lei Liang,&nbsp;Wen Jin,&nbsp;Yan-Ping Wu,&nbsp;Rong-Rong He,&nbsp;Yi-Fang Li","doi":"10.1038/s41418-024-01351-0","DOIUrl":"10.1038/s41418-024-01351-0","url":null,"abstract":"Ferroptosis holds significant potential for application in cancer therapy. However, ferroptosis inducers are not cell-specific and can cause phospholipid peroxidation in both tumor and non-tumor cells. This limitation greatly restricts the use of ferroptosis therapy as a safe and effective anticancer strategy. Our previous study demonstrated that macrophages can engulf ferroptotic cells through Toll-like receptor 2 (TLR2). Despite this advancement, the precise mechanism by which phospholipid peroxidation in macrophages affects their phagocytotic capability during treatment of tumors with ferroptotic agents is still unknown. Here, we utilized flow sorting combined with redox phospholipidomics to determine that phospholipid peroxidation in tumor microenvironment (TME) macrophages impaired the macrophages ability to eliminate ferroptotic tumor cells by phagocytosis, ultimately fostering tumor resistance to ferroptosis therapy. Mechanistically, the accumulation of phospholipid peroxidation in the macrophage endoplasmic reticulum (ER) repressed TLR2 trafficking to the plasma membrane and caused its retention in the ER by disrupting the interaction between TLR2 and its chaperone CNPY3. Subsequently, this ER-retained TLR2 recruited E3 ligase MARCH6 and initiated the proteasome-dependent degradation. Using redox phospholipidomics, we identified 1-steaoryl-2-15-HpETE-sn-glycero-3-phosphatidylethanolamine (SAPE-OOH) as the crucial mediator of these effects. Conclusively, our discovery elucidates a novel molecular mechanism underlying macrophage phospholipid peroxidation-induced tumor resistance to ferroptosis therapy and highlights the TLR2-MARCH6 axis as a potential therapeutic target for cancer therapy.","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 9","pages":"1184-1201"},"PeriodicalIF":13.7,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141892928","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
Correction to: Brf1 loss and not overexpression disrupts tissues homeostasis in the intestine, liver and pancreas 更正为Brf1缺失而非过度表达会破坏肠道、肝脏和胰腺的组织平衡。
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-02 DOI: 10.1038/s41418-024-01344-z
Dritan Liko, Louise Mitchell, Kirsteen J. Campbell, Rachel A. Ridgway, Carolyn Jones, Kate Dudek, Ayala King, Sheila Bryson, David Stevenson, Karen Blyth, Douglas Strathdee, Jennifer P. Morton, Thomas G. Bird, John R. P. Knight, Anne E. Willis, Owen J. Sansom
{"title":"Correction to: Brf1 loss and not overexpression disrupts tissues homeostasis in the intestine, liver and pancreas","authors":"Dritan Liko,&nbsp;Louise Mitchell,&nbsp;Kirsteen J. Campbell,&nbsp;Rachel A. Ridgway,&nbsp;Carolyn Jones,&nbsp;Kate Dudek,&nbsp;Ayala King,&nbsp;Sheila Bryson,&nbsp;David Stevenson,&nbsp;Karen Blyth,&nbsp;Douglas Strathdee,&nbsp;Jennifer P. Morton,&nbsp;Thomas G. Bird,&nbsp;John R. P. Knight,&nbsp;Anne E. Willis,&nbsp;Owen J. Sansom","doi":"10.1038/s41418-024-01344-z","DOIUrl":"10.1038/s41418-024-01344-z","url":null,"abstract":"","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 9","pages":"1235-1237"},"PeriodicalIF":13.7,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41418-024-01344-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141878449","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
TIN2 modulates FOXO1 mitochondrial shuttling to enhance oxidative stress-induced apoptosis in retinal pigment epithelium under hyperglycemia TIN2调节FOXO1线粒体穿梭,以增强高血糖状态下氧化应激诱导的视网膜色素上皮细胞凋亡。
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-30 DOI: 10.1038/s41418-024-01349-8
Shimei Chen, Dandan Sun, Shuchang Zhang, Li Xu, Ning Wang, Huiming Li, Xun Xu, Fang Wei
Progressive dysfunction of the retinal pigment epithelium (RPE) and the adjacent photoreceptor cells in the outer retina plays a pivotal role in the pathogenesis of diabetic retinopathy (DR). Here, we observed a marked increase in oxidative stress-induced apoptosis in parallel with higher expression of telomeric protein TIN2 in RPE cells under hyperglycemia in vivo and in vitro. Delving deeper, we confirm that high glucose-induced elevation of mitochondria-localized TIN2 compromises mitochondrial activity and weakens the intrinsic antioxidant defense, thereby leading to the activation of mitochondria-dependent apoptotic pathways. Mechanistically, mitochondrial TIN2 promotes the phosphorylation of FOXO1 and its relocation to the mitochondria. Such translocation of transcription factor FOXO1 not only promotes its binding to the D-loop region of mitochondrial DNA—resulting in the inhibition of mitochondrial respiration—but also hampers its availability to nuclear target DNA, thereby undermining the intrinsic antioxidant defense. Moreover, TIN2 knockdown effectively mitigates oxidative-induced apoptosis in diabetic mouse RPE by preserving mitochondrial homeostasis, which concurrently prevents secondary photoreceptor damage. Our study proposes the potential of TIN2 as a promising molecular target for therapeutic interventions for diabetic retinopathy, which emphasizes the potential significance of telomeric proteins in the regulation of metabolism and mitochondrial function.
在糖尿病视网膜病变(DR)的发病机制中,视网膜外层的视网膜色素上皮细胞(RPE)和邻近的感光细胞的进行性功能障碍起着关键作用。在这里,我们观察到在体内和体外高血糖条件下,RPE 细胞中端粒蛋白 TIN2 的表达增加的同时,氧化应激诱导的细胞凋亡也明显增加。深入研究后,我们证实高血糖诱导的线粒体定位 TIN2 的升高损害了线粒体活性,削弱了内在抗氧化防御能力,从而导致线粒体依赖性凋亡通路的激活。从机制上讲,线粒体 TIN2 会促进 FOXO1 的磷酸化并将其转移到线粒体。转录因子 FOXO1 的这种转位不仅会促进其与线粒体 DNA 的 D-loop 区域结合,从而抑制线粒体呼吸,而且还会阻碍其与核靶 DNA 的结合,从而破坏内在的抗氧化防御。此外,TIN2基因敲除可通过维持线粒体平衡有效缓解氧化诱导的糖尿病小鼠RPE细胞凋亡,同时防止继发性感光细胞损伤。我们的研究提出了TIN2作为糖尿病视网膜病变治疗干预分子靶点的潜力,这强调了端粒蛋白在调节新陈代谢和线粒体功能方面的潜在意义。用 BioRender ( https://www.biorender.com/ ) 制作。
{"title":"TIN2 modulates FOXO1 mitochondrial shuttling to enhance oxidative stress-induced apoptosis in retinal pigment epithelium under hyperglycemia","authors":"Shimei Chen,&nbsp;Dandan Sun,&nbsp;Shuchang Zhang,&nbsp;Li Xu,&nbsp;Ning Wang,&nbsp;Huiming Li,&nbsp;Xun Xu,&nbsp;Fang Wei","doi":"10.1038/s41418-024-01349-8","DOIUrl":"10.1038/s41418-024-01349-8","url":null,"abstract":"Progressive dysfunction of the retinal pigment epithelium (RPE) and the adjacent photoreceptor cells in the outer retina plays a pivotal role in the pathogenesis of diabetic retinopathy (DR). Here, we observed a marked increase in oxidative stress-induced apoptosis in parallel with higher expression of telomeric protein TIN2 in RPE cells under hyperglycemia in vivo and in vitro. Delving deeper, we confirm that high glucose-induced elevation of mitochondria-localized TIN2 compromises mitochondrial activity and weakens the intrinsic antioxidant defense, thereby leading to the activation of mitochondria-dependent apoptotic pathways. Mechanistically, mitochondrial TIN2 promotes the phosphorylation of FOXO1 and its relocation to the mitochondria. Such translocation of transcription factor FOXO1 not only promotes its binding to the D-loop region of mitochondrial DNA—resulting in the inhibition of mitochondrial respiration—but also hampers its availability to nuclear target DNA, thereby undermining the intrinsic antioxidant defense. Moreover, TIN2 knockdown effectively mitigates oxidative-induced apoptosis in diabetic mouse RPE by preserving mitochondrial homeostasis, which concurrently prevents secondary photoreceptor damage. Our study proposes the potential of TIN2 as a promising molecular target for therapeutic interventions for diabetic retinopathy, which emphasizes the potential significance of telomeric proteins in the regulation of metabolism and mitochondrial function.","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 11","pages":"1487-1505"},"PeriodicalIF":13.7,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854869","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
TOX2 nuclear-cytosol translocation is linked to leukemogenesis of acute T-cell leukemia by repressing TIM3 transcription TOX2核-胞浆转位通过抑制TIM3转录与急性T细胞白血病的白血病发生有关。
IF 13.7 1区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-07-30 DOI: 10.1038/s41418-024-01352-z
Anzhou Li, Junbao Zhang, Liangping Zhan, Xiufeng Liu, Xiliang Zeng, Qian Zhu, Zifeng Wang, Jiang Li
Nuclear factors TOX and TOX2 upregulate TIM3 expression and lead to T-cell exhaustion in malignancies. Here, we demonstrate two distinct TIM3 expression patterns (high & low) with high TOX and TOX2 levels in T-cell acute lymphoblastic leukemia (T-ALL) specimens and cell lines. However, the mechanisms regulated by TOX and TIM3 signaling in leukemogenesis are unclear. We found that TOX and TOX2 proteins each directly upregulated HAVCR2 transcription, while the cellular localization of TOX2 was different in Jurkat and MOLT3 cells (nucleus) and lymphoblastic cell T2 and normal T cells (cytoplasm). Nuclear TOX and TOX2 formed a protein complex and repressed HAVCR2 promoter activity by recruiting transcriptional corepressor LCOR and deacetylase HDAC3. The nuclear-cytosol translocation of TOX2 was deacetylation-dependent and cooperatively mediated by deacetylase Sirt1 and kinase TBK1. Radiation damage induced TOX2 nuclear translocation and decreased Sirt1, TIM3, and caspase 1 expression in normal T cells. Accordingly, knockdown of TOX, TOX2 or LCOR; HDAC3 inhibition; or TIM3 overexpression induced Jurkat cell apoptosis in vitro and slow growth in vivo. Thus, our findings demonstrate a novel regulatory mechanism involving TOX-TOX2 and the TIM3 pathway in the leukemogenesis of T-ALL.
核因子 TOX 和 TOX2 能上调 TIM3 的表达,导致恶性肿瘤中 T 细胞衰竭。在这里,我们展示了T细胞急性淋巴细胞白血病(T-ALL)标本和细胞系中两种不同的TIM3表达模式(高和低),以及高水平的TOX和TOX2。然而,TOX 和 TIM3 信号在白血病发生过程中的调控机制尚不清楚。我们发现,TOX 和 TOX2 蛋白各自直接上调 HAVCR2 的转录,而 TOX2 在 Jurkat 和 MOLT3 细胞(细胞核)以及淋巴母细胞 T2 和正常 T 细胞(细胞质)中的细胞定位不同。核TOX和TOX2形成蛋白复合物,通过招募转录核心抑制因子LCOR和去乙酰化酶HDAC3抑制HAVCR2启动子的活性。TOX2的核-胞浆转位依赖于去乙酰化,并由去乙酰化酶Sirt1和激酶TBK1协同介导。辐射损伤诱导了TOX2的核转位,并降低了正常T细胞中Sirt1、TIM3和caspase 1的表达。因此,体外敲除 TOX、TOX2 或 LCOR;抑制 HDAC3 或 TIM3 过表达可诱导 Jurkat 细胞凋亡,并减缓体内生长。因此,我们的研究结果证明了TOX-TOX2和TIM3通路在T-ALL白血病发生过程中的新型调控机制。
{"title":"TOX2 nuclear-cytosol translocation is linked to leukemogenesis of acute T-cell leukemia by repressing TIM3 transcription","authors":"Anzhou Li,&nbsp;Junbao Zhang,&nbsp;Liangping Zhan,&nbsp;Xiufeng Liu,&nbsp;Xiliang Zeng,&nbsp;Qian Zhu,&nbsp;Zifeng Wang,&nbsp;Jiang Li","doi":"10.1038/s41418-024-01352-z","DOIUrl":"10.1038/s41418-024-01352-z","url":null,"abstract":"Nuclear factors TOX and TOX2 upregulate TIM3 expression and lead to T-cell exhaustion in malignancies. Here, we demonstrate two distinct TIM3 expression patterns (high &amp; low) with high TOX and TOX2 levels in T-cell acute lymphoblastic leukemia (T-ALL) specimens and cell lines. However, the mechanisms regulated by TOX and TIM3 signaling in leukemogenesis are unclear. We found that TOX and TOX2 proteins each directly upregulated HAVCR2 transcription, while the cellular localization of TOX2 was different in Jurkat and MOLT3 cells (nucleus) and lymphoblastic cell T2 and normal T cells (cytoplasm). Nuclear TOX and TOX2 formed a protein complex and repressed HAVCR2 promoter activity by recruiting transcriptional corepressor LCOR and deacetylase HDAC3. The nuclear-cytosol translocation of TOX2 was deacetylation-dependent and cooperatively mediated by deacetylase Sirt1 and kinase TBK1. Radiation damage induced TOX2 nuclear translocation and decreased Sirt1, TIM3, and caspase 1 expression in normal T cells. Accordingly, knockdown of TOX, TOX2 or LCOR; HDAC3 inhibition; or TIM3 overexpression induced Jurkat cell apoptosis in vitro and slow growth in vivo. Thus, our findings demonstrate a novel regulatory mechanism involving TOX-TOX2 and the TIM3 pathway in the leukemogenesis of T-ALL.","PeriodicalId":9731,"journal":{"name":"Cell Death and Differentiation","volume":"31 11","pages":"1506-1518"},"PeriodicalIF":13.7,"publicationDate":"2024-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41418-024-01352-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141854870","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 and Differentiation
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号: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