首页 > 最新文献

Cell Death & Disease最新文献

英文 中文
DDX5 deficiency drives non-canonical NF-κB activation and NRF2 expression, influencing sorafenib response and hepatocellular carcinoma progression. DDX5 缺乏会导致非典型 NF-κB 激活和 NRF2 表达,从而影响索拉非尼反应和肝细胞癌的进展。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-09 DOI: 10.1038/s41419-024-06977-z
Zhili Li, Woojun Kim, Sagar Utturkar, Bingyu Yan, Nadia Atallah Lanman, Bennett D Elzey, Majid Kazemian, Yoon Yeo, Ourania Andrisani

In advanced hepatocellular carcinoma (HCC), RNA helicase DDX5 regulates the Wnt/β-catenin-ferroptosis axis, influencing the efficacy of the multi-tyrosine kinase inhibitor (mTKI) sorafenib. DDX5 inhibits Wnt/β-catenin signaling, preventing sorafenib-induced ferroptosis escape. Sorafenib/mTKIs reduce DDX5 expression, correlating with poor patient survival post-sorafenib treatment. Notably, DDX5-knockout in HCC cells activates Wnt/β-catenin signaling persistently. Herein, we investigate the mechanistic impact of Wnt/β-catenin activation resulting from DDX5 downregulation in the progression and treatment of HCC. RNAseq analyses identified shared genes repressed by DDX5 and upregulated by sorafenib, including Wnt signaling genes, NF-κB-inducing kinase (NIK) essential for non-canonical NF-κB (p52/RelB) activation, and cytoprotective transcription factor NRF2. We demonstrate, Wnt/β-catenin activation induced NIK transcription, leading to non-canonical NF-κB activation, which subsequently mediated NRF2 transcription. Additionally, DDX5 deficiency extended NRF2 protein half-life by inactivating KEAP1 through p62/SQSTM1 stabilization. In a preclinical HCC mouse model, NRF2 knockdown or DDX5 overexpression restricted tumor growth upon sorafenib treatment, via induction of ferroptosis. Importantly, DDX5-knockout HCC cells exhibited elevated expression of Wnt signaling genes, NIK, p52/RelB, and NRF2-regulated genes, regardless of sorafenib treatment. Transcriptomic analyses of HCCs from TCGA and the Stelic Animal Model (STAM) of non-alcoholic steatohepatitis revealed elevated expression of these interconnected pathways in the context of DDX5 downregulation. In conclusion, DDX5 deficiency triggers Wnt/β-catenin signaling, promoting p52/RelB and NRF2 activation, thereby enabling ferroptosis evasion upon sorafenib treatment. Similarly, independent of sorafenib, DDX5 deficiency in liver tumors enhances activation and gene expression of these interconnected pathways, underscoring the clinical relevance of DDX5 deficiency in HCC progression and therapeutic response.

在晚期肝细胞癌(HCC)中,RNA螺旋酶DDX5调控Wnt/β-catenin-铁突变轴,影响多酪氨酸激酶抑制剂(mTKI)索拉非尼的疗效。DDX5 可抑制 Wnt/β-catenin 信号转导,防止索拉非尼诱导的铁突变逃逸。索拉非尼/mTKIs会降低DDX5的表达,这与索拉非尼治疗后患者生存率低有关。值得注意的是,HCC细胞中DDX5基因敲除会持续激活Wnt/β-catenin信号。在此,我们研究了DDX5下调导致的Wnt/β-catenin激活对HCC进展和治疗的机理影响。RNAseq分析发现了被DDX5抑制、被索拉非尼上调的共有基因,包括Wnt信号基因、非典型NF-κB(p52/RelB)激活所必需的NF-κB诱导激酶(NIK)以及细胞保护转录因子NRF2。我们发现,Wnt/β-catenin活化诱导NIK转录,导致非经典NF-κB活化,进而介导NRF2转录。此外,缺乏 DDX5 会通过 p62/SQSTM1 的稳定使 KEAP1 失活,从而延长 NRF2 蛋白的半衰期。在临床前 HCC 小鼠模型中,NRF2 基因敲除或 DDX5 基因过表达通过诱导铁变态反应限制了索拉非尼治疗后的肿瘤生长。重要的是,无论索拉非尼治疗与否,DDX5敲除的HCC细胞都表现出Wnt信号基因、NIK、p52/RelB和NRF2调控基因的表达升高。对来自 TCGA 和非酒精性脂肪性肝炎 Stelic 动物模型(STAM)的 HCC 进行转录组学分析发现,在 DDX5 下调的情况下,这些相互关联的通路的表达也会升高。总之,DDX5缺乏会触发Wnt/β-catenin信号转导,促进p52/RelB和NRF2的活化,从而使索拉非尼治疗后的铁变态反应得以逃避。同样,与索拉非尼无关,肝脏肿瘤中的DDX5缺乏也会增强这些相互关联的通路的激活和基因表达,从而强调了DDX5缺乏在HCC进展和治疗反应中的临床意义。
{"title":"DDX5 deficiency drives non-canonical NF-κB activation and NRF2 expression, influencing sorafenib response and hepatocellular carcinoma progression.","authors":"Zhili Li, Woojun Kim, Sagar Utturkar, Bingyu Yan, Nadia Atallah Lanman, Bennett D Elzey, Majid Kazemian, Yoon Yeo, Ourania Andrisani","doi":"10.1038/s41419-024-06977-z","DOIUrl":"10.1038/s41419-024-06977-z","url":null,"abstract":"<p><p>In advanced hepatocellular carcinoma (HCC), RNA helicase DDX5 regulates the Wnt/β-catenin-ferroptosis axis, influencing the efficacy of the multi-tyrosine kinase inhibitor (mTKI) sorafenib. DDX5 inhibits Wnt/β-catenin signaling, preventing sorafenib-induced ferroptosis escape. Sorafenib/mTKIs reduce DDX5 expression, correlating with poor patient survival post-sorafenib treatment. Notably, DDX5-knockout in HCC cells activates Wnt/β-catenin signaling persistently. Herein, we investigate the mechanistic impact of Wnt/β-catenin activation resulting from DDX5 downregulation in the progression and treatment of HCC. RNAseq analyses identified shared genes repressed by DDX5 and upregulated by sorafenib, including Wnt signaling genes, NF-κB-inducing kinase (NIK) essential for non-canonical NF-κB (p52/RelB) activation, and cytoprotective transcription factor NRF2. We demonstrate, Wnt/β-catenin activation induced NIK transcription, leading to non-canonical NF-κB activation, which subsequently mediated NRF2 transcription. Additionally, DDX5 deficiency extended NRF2 protein half-life by inactivating KEAP1 through p62/SQSTM1 stabilization. In a preclinical HCC mouse model, NRF2 knockdown or DDX5 overexpression restricted tumor growth upon sorafenib treatment, via induction of ferroptosis. Importantly, DDX5-knockout HCC cells exhibited elevated expression of Wnt signaling genes, NIK, p52/RelB, and NRF2-regulated genes, regardless of sorafenib treatment. Transcriptomic analyses of HCCs from TCGA and the Stelic Animal Model (STAM) of non-alcoholic steatohepatitis revealed elevated expression of these interconnected pathways in the context of DDX5 downregulation. In conclusion, DDX5 deficiency triggers Wnt/β-catenin signaling, promoting p52/RelB and NRF2 activation, thereby enabling ferroptosis evasion upon sorafenib treatment. Similarly, independent of sorafenib, DDX5 deficiency in liver tumors enhances activation and gene expression of these interconnected pathways, underscoring the clinical relevance of DDX5 deficiency in HCC progression and therapeutic response.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11315975/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911980","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
Cyld restrains the hyperactivation of synovial fibroblasts in inflammatory arthritis by regulating the TAK1/IKK2 signaling axis. Cyld 通过调节 TAK1/IKK2 信号轴抑制炎症性关节炎中滑膜成纤维细胞的过度激活。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-09 DOI: 10.1038/s41419-024-06966-2
Vagelis Rinotas, Kalliopi Iliaki, Lydia Pavlidi, Theodore Meletakos, George Mosialos, Marietta Armaka

TNF is a potent cytokine known for its involvement in physiology and pathology. In Rheumatoid Arthritis (RA), persistent TNF signals cause aberrant activation of synovial fibroblasts (SFs), the resident cells crucially involved in the inflammatory and destructive responses of the affected synovial membrane. However, the molecular switches that control the pathogenic activation of SFs remain poorly defined. Cyld is a major component of deubiquitination (DUB) machinery regulating the signaling responses towards survival/inflammation and programmed necrosis that induced by cytokines, growth factors and microbial products. Herein, we follow functional genetic approaches to understand how Cyld affects arthritogenic TNF signaling in SFs. We demonstrate that in spontaneous and induced RA models, SF-Cyld DUB deficiency deteriorates arthritic phenotypes due to increased levels of chemokines, adhesion receptors and bone-degrading enzymes generated by mutant SFs. Mechanistically, Cyld serves to restrict the TNF-induced hyperactivation of SFs by limiting Tak1-mediated signaling, and, therefore, leading to supervised NF-κB and JNK activity. However, Cyld is not critically involved in the regulation of TNF-induced death of SFs. Our results identify SF-Cyld as a regulator of TNF-mediated arthritis and inform the signaling landscape underpinning the SF responses.

TNF 是一种强效细胞因子,因其参与生理学和病理学而闻名。在类风湿性关节炎(RA)中,持续的 TNF 信号会导致滑膜成纤维细胞(SFs)异常活化,SFs 是一种驻留细胞,在受影响滑膜的炎症和破坏性反应中起着至关重要的作用。然而,控制滑膜成纤维细胞致病性活化的分子开关仍未明确。Cyld 是去泛素化(DUB)机制的主要组成部分,它调节着细胞因子、生长因子和微生物产物诱导的生存/炎症和程序性坏死信号反应。在此,我们采用功能基因方法来了解 Cyld 如何影响 SFs 中的致关节炎 TNF 信号转导。我们证明,在自发性和诱导性 RA 模型中,由于突变 SFs 产生的趋化因子、粘附受体和骨降解酶水平升高,SF-Cyld DUB 缺乏会使关节炎表型恶化。从机理上讲,Cyld通过限制Tak1介导的信号传导来限制TNF诱导的SFs过度激活,从而导致NF-κB和JNK活性受到监督。然而,Cyld并没有关键性地参与TNF诱导的SFs死亡调控。我们的研究结果确定了 SF-Cyld 是 TNF 介导的关节炎的调控因子,并为 SF 反应的信号传导机制提供了信息。
{"title":"Cyld restrains the hyperactivation of synovial fibroblasts in inflammatory arthritis by regulating the TAK1/IKK2 signaling axis.","authors":"Vagelis Rinotas, Kalliopi Iliaki, Lydia Pavlidi, Theodore Meletakos, George Mosialos, Marietta Armaka","doi":"10.1038/s41419-024-06966-2","DOIUrl":"10.1038/s41419-024-06966-2","url":null,"abstract":"<p><p>TNF is a potent cytokine known for its involvement in physiology and pathology. In Rheumatoid Arthritis (RA), persistent TNF signals cause aberrant activation of synovial fibroblasts (SFs), the resident cells crucially involved in the inflammatory and destructive responses of the affected synovial membrane. However, the molecular switches that control the pathogenic activation of SFs remain poorly defined. Cyld is a major component of deubiquitination (DUB) machinery regulating the signaling responses towards survival/inflammation and programmed necrosis that induced by cytokines, growth factors and microbial products. Herein, we follow functional genetic approaches to understand how Cyld affects arthritogenic TNF signaling in SFs. We demonstrate that in spontaneous and induced RA models, SF-Cyld DUB deficiency deteriorates arthritic phenotypes due to increased levels of chemokines, adhesion receptors and bone-degrading enzymes generated by mutant SFs. Mechanistically, Cyld serves to restrict the TNF-induced hyperactivation of SFs by limiting Tak1-mediated signaling, and, therefore, leading to supervised NF-κB and JNK activity. However, Cyld is not critically involved in the regulation of TNF-induced death of SFs. Our results identify SF-Cyld as a regulator of TNF-mediated arthritis and inform the signaling landscape underpinning the SF responses.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11316070/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141911978","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
Regulation of Zfp36 by ISGF3 and MK2 restricts the expression of inflammatory cytokines during necroptosis stimulation. ISGF3 和 MK2 对 Zfp36 的调控限制了坏死刺激过程中炎性细胞因子的表达。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41419-024-06964-4
Sahil Yadav, Rayan El Hamra, Norah A Alturki, Ardeshir Ariana, Avni Bhan, Kate Hurley, Matthias Gaestel, Perry J Blackshear, Alexandre Blais, Subash Sad

Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNβ, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNβ-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNβ, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.

坏死体在 TLR 或细胞因子受体信号传导后被激活,导致细胞坏死,其特点是细胞膜破裂,细胞内内容物随之释放到细胞外环境中。虽然坏死细胞增多会加剧各种炎症性疾病,但炎症反应的调控机制尚不清楚。我们的研究表明,巨噬细胞的坏死体激活会导致各种通路(包括丝裂原活化蛋白激酶(MAPK)级联)的上调,从而导致炎症反应的升高以及随之而来的多种细胞因子和趋化因子的表达。炎症反应上调的程序设计发生在坏死体活化的早期阶段,与细胞死亡无关,但取决于受体相互作用蛋白激酶-1(RipK1)的活化。有趣的是,坏死体的激活也会导致 IFNβ 的上调,而 IFNβ 又会通过抑制 MAPK 信号传导和 Zfp36 的上调对炎症反应的维持产生抑制作用。干扰素诱导基因因子-3(ISGF3)的激活会导致 ZFP36(TTP)的表达,ZFP36 通过识别各种炎症细胞因子和趋化因子 3'UTR 中富含的 AU 元素,诱导其 mRNA 的转录后降解。此外,ZFP-36 还能抑制 IFNβ,但不能抑制 TNFα 诱导的坏死。总之,这些结果揭示了 IFNβ(一种促炎细胞因子)诱导 ZFP-36 表达的分子机制,ZFP-36 反过来又抑制坏死并阻止炎症反应的维持。
{"title":"Regulation of Zfp36 by ISGF3 and MK2 restricts the expression of inflammatory cytokines during necroptosis stimulation.","authors":"Sahil Yadav, Rayan El Hamra, Norah A Alturki, Ardeshir Ariana, Avni Bhan, Kate Hurley, Matthias Gaestel, Perry J Blackshear, Alexandre Blais, Subash Sad","doi":"10.1038/s41419-024-06964-4","DOIUrl":"10.1038/s41419-024-06964-4","url":null,"abstract":"<p><p>Necrosome activation following TLR- or cytokine receptor-signaling results in cell death by necroptosis which is characterized by the rupture of cell membranes and the consequent release of intracellular contents to the extracellular milieu. While necroptosis exacerbates various inflammatory diseases, the mechanisms through which the inflammatory responses are regulated are not clear. We show that the necrosome activation of macrophages results in an upregulation of various pathways, including the mitogen-activated protein kinase (MAPK) cascade, which results in an elevation of the inflammatory response and consequent expression of several cytokines and chemokines. Programming for this upregulation of inflammatory response occurs during the early phase of necrosome activation and proceeds independently of cell death but depends on the activation of the receptor-interacting protein kinase-1 (RipK1). Interestingly, necrosome activation also results in an upregulation of IFNβ, which in turn exerts an inhibitory effect on the maintenance of inflammatory response through the repression of MAPK-signaling and an upregulation of Zfp36. Activation of the interferon-induced gene factor-3 (ISGF3) results in the expression of ZFP36 (TTP), which induces the post-transcriptional degradation of mRNAs of various inflammatory cytokines and chemokines through the recognition of AU-rich elements in their 3'UTR. Furthermore, ZFP-36 inhibits IFNβ-, but not TNFα- induced necroptosis. Overall, these results reveal the molecular mechanism through which IFNβ, a pro-inflammatory cytokine, induces the expression of ZFP-36, which in turn inhibits necroptosis and halts the maintenance of the inflammatory response.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310327/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906020","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
ALYREF promotes the metastasis of nasopharyngeal carcinoma by increasing the stability of NOTCH1 mRNA. ALYREF 通过增加 NOTCH1 mRNA 的稳定性促进鼻咽癌的转移。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41419-024-06959-1
Yanan Jin, Jijin Yao, Jianchang Fu, Qitao Huang, Yilin Luo, Yafei You, Wangjian Zhang, Qian Zhong, Tianliang Xia, Liangping Xia

Approximately 70% of treatment failures in nasopharyngeal carcinoma (NPC) patients are attributed to distant metastasis, yet the underlying mechanisms remain unclear. RNA 5-methylcytosine (m5C) is an emerging regulatory modification that controls gene expression and plays a critical role in tumor progression. However, there is little information on the potential roles of RNA m5C modification in NPC metastasis. In this study, we found that the m5C reader Aly/REF export factor (ALYREF) is significantly upregulated in NPC, whereby its high expression is associated with metastasis and poor prognosis. ALYREF overexpression was found to promote tumor metastasis of NPC cells in vitro and in vivo. Mechanistically, m5C-modified NOTCH1 mRNA was identified as a target of ALYREF. Moreover, ALYREF was found to upregulate NOTCH1 expression by enhancing its RNA stability in an m5C modification-dependent manner, thereby promoting the activation of the NOTCH signaling pathway and facilitating NPC metastasis. Overall, our data reveal the crucial role of ALYREF in NPC metastasis and provide a potential therapeutic target for NPC.

鼻咽癌(NPC)患者中约有 70% 的治疗失败归因于远处转移,但其潜在机制仍不清楚。RNA 5-甲基胞嘧啶(m5C)是一种新出现的调控修饰,可控制基因表达并在肿瘤进展中发挥关键作用。然而,关于 RNA m5C 修饰在鼻咽癌转移中的潜在作用的信息却很少。在本研究中,我们发现 m5C 阅读器 Aly/REF 导出因子(ALYREF)在鼻咽癌中显著上调,而其高表达与转移和不良预后相关。研究发现,ALYREF 的过表达可促进鼻咽癌细胞在体外和体内的转移。从机理上讲,m5C修饰的NOTCH1 mRNA被确定为ALYREF的靶标。此外,研究还发现,ALYREF 通过提高 NOTCH1 的 RNA 稳定性,以 m5C 修饰依赖性的方式上调 NOTCH1 的表达,从而促进 NOTCH 信号通路的激活并促进鼻咽癌的转移。总之,我们的数据揭示了ALYREF在鼻咽癌转移中的关键作用,并为鼻咽癌提供了一个潜在的治疗靶点。
{"title":"ALYREF promotes the metastasis of nasopharyngeal carcinoma by increasing the stability of NOTCH1 mRNA.","authors":"Yanan Jin, Jijin Yao, Jianchang Fu, Qitao Huang, Yilin Luo, Yafei You, Wangjian Zhang, Qian Zhong, Tianliang Xia, Liangping Xia","doi":"10.1038/s41419-024-06959-1","DOIUrl":"10.1038/s41419-024-06959-1","url":null,"abstract":"<p><p>Approximately 70% of treatment failures in nasopharyngeal carcinoma (NPC) patients are attributed to distant metastasis, yet the underlying mechanisms remain unclear. RNA 5-methylcytosine (m5C) is an emerging regulatory modification that controls gene expression and plays a critical role in tumor progression. However, there is little information on the potential roles of RNA m5C modification in NPC metastasis. In this study, we found that the m5C reader Aly/REF export factor (ALYREF) is significantly upregulated in NPC, whereby its high expression is associated with metastasis and poor prognosis. ALYREF overexpression was found to promote tumor metastasis of NPC cells in vitro and in vivo. Mechanistically, m5C-modified NOTCH1 mRNA was identified as a target of ALYREF. Moreover, ALYREF was found to upregulate NOTCH1 expression by enhancing its RNA stability in an m5C modification-dependent manner, thereby promoting the activation of the NOTCH signaling pathway and facilitating NPC metastasis. Overall, our data reveal the crucial role of ALYREF in NPC metastasis and provide a potential therapeutic target for NPC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310353/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906053","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
Potential of GSPT1 as a novel target for glioblastoma therapy. GSPT1 作为胶质母细胞瘤治疗新靶点的潜力。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41419-024-06967-1
Takashi Sasayama, Takeshi Hamada, Kazuhiro Tanaka, Hiroaki Nagashima, Shunsuke Yamanishi, Takehiko Ueyama

Glioblastoma is the most common malignant brain tumor in adults, the survival rate of which has not significantly improved over the past three decades. Therefore, there is an urgent need to develop novel treatment modalities. We previously reported that G1 to S phase transition 1 (GSPT1) depletion induces delayed cell cycle in primary astrocytes. Herein, we examined the potential of GSPT1 as a novel target for glioblastoma therapy. CC-885, a cereblon modulator that degrades GSPT1 by bridging GSPT1 to the CRL4 E3 ubiquitin ligase complex, was administered to nude mice with transplanted brain tumors of U87 glioblastoma cells. The survival period was significantly longer in CC-885 treated mice than in control mice. Furthermore, we generated GSPT1-knockout (KO) U87 cells and GSPT1-KO U87 cells with stable overexpression of FLAG-tagged GSPT1 (Rescued GSPT1-KO). Mice with transplanted GSPT1-KO U87 cells and Rescued GSPT1-KO U87 cells showed significantly longer and similar survival periods, respectively, as those with wild-type (WT) U87 cells. GSPT1-KO U87 cells showed enhanced apoptosis, detected by cleaved PARP1, compared to WT U87 cells. Brain tumors with transplantation of GSPT1-KO U87 cells also showed enhanced apoptosis compared to those with transplantation of WT and Rescued GSPT1-KO U87 cells. GSPT1 expression was confirmed in patients with glioblastoma. However, the clinical study using 87 glioblastoma samples showed that GSPT1 mRNA levels were not associated with overall survival. Taken together, we propose that GSPT1 is an essential protein for glioblastoma growth, but not its malignant characteristics, and that GSPT1 is a potential target for developing glioblastoma therapeutics.

胶质母细胞瘤是成人中最常见的恶性脑肿瘤,其存活率在过去三十年中没有明显改善。因此,迫切需要开发新的治疗模式。我们以前曾报道过,G1 到 S 期转变 1(GSPT1)耗竭会诱导原发性星形胶质细胞的细胞周期延迟。在此,我们研究了GSPT1作为胶质母细胞瘤治疗新靶点的潜力。CC-885是一种脑龙调节剂,它通过将GSPT1与CRL4 E3泛素连接酶复合物连接来降解GSPT1。接受CC-885治疗的小鼠的存活期明显长于对照组小鼠。此外,我们还生成了GSPT1基因敲除(KO)的U87细胞和稳定过表达FLAG标记的GSPT1的GSPT1-KO U87细胞(Rescued GSPT1-KO)。移植了GSPT1-KO U87细胞和Rescued GSPT1-KO U87细胞的小鼠存活期明显更长,与移植了野生型(WT)U87细胞的小鼠存活期相似。与WT U87细胞相比,GSPT1-KO U87细胞通过PARP1裂解检测到的细胞凋亡增强。移植了 GSPT1-KO U87 细胞的脑肿瘤与移植了 WT 和 Rescued GSPT1-KO U87 细胞的脑肿瘤相比,也显示出更强的细胞凋亡能力。在胶质母细胞瘤患者中证实了 GSPT1 的表达。然而,使用 87 例胶质母细胞瘤样本进行的临床研究显示,GSPT1 mRNA 水平与总生存率无关。综上所述,我们认为 GSPT1 是胶质母细胞瘤生长的必需蛋白,但不是其恶性特征的必需蛋白,GSPT1 是开发胶质母细胞瘤疗法的潜在靶点。
{"title":"Potential of GSPT1 as a novel target for glioblastoma therapy.","authors":"Takashi Sasayama, Takeshi Hamada, Kazuhiro Tanaka, Hiroaki Nagashima, Shunsuke Yamanishi, Takehiko Ueyama","doi":"10.1038/s41419-024-06967-1","DOIUrl":"10.1038/s41419-024-06967-1","url":null,"abstract":"<p><p>Glioblastoma is the most common malignant brain tumor in adults, the survival rate of which has not significantly improved over the past three decades. Therefore, there is an urgent need to develop novel treatment modalities. We previously reported that G1 to S phase transition 1 (GSPT1) depletion induces delayed cell cycle in primary astrocytes. Herein, we examined the potential of GSPT1 as a novel target for glioblastoma therapy. CC-885, a cereblon modulator that degrades GSPT1 by bridging GSPT1 to the CRL4 E3 ubiquitin ligase complex, was administered to nude mice with transplanted brain tumors of U87 glioblastoma cells. The survival period was significantly longer in CC-885 treated mice than in control mice. Furthermore, we generated GSPT1-knockout (KO) U87 cells and GSPT1-KO U87 cells with stable overexpression of FLAG-tagged GSPT1 (Rescued GSPT1-KO). Mice with transplanted GSPT1-KO U87 cells and Rescued GSPT1-KO U87 cells showed significantly longer and similar survival periods, respectively, as those with wild-type (WT) U87 cells. GSPT1-KO U87 cells showed enhanced apoptosis, detected by cleaved PARP1, compared to WT U87 cells. Brain tumors with transplantation of GSPT1-KO U87 cells also showed enhanced apoptosis compared to those with transplantation of WT and Rescued GSPT1-KO U87 cells. GSPT1 expression was confirmed in patients with glioblastoma. However, the clinical study using 87 glioblastoma samples showed that GSPT1 mRNA levels were not associated with overall survival. Taken together, we propose that GSPT1 is an essential protein for glioblastoma growth, but not its malignant characteristics, and that GSPT1 is a potential target for developing glioblastoma therapeutics.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310507/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906018","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
Age-related retinal degeneration resulting from the deletion of Shp2 tyrosine phosphatase in photoreceptor neurons. 感光神经元中 Shp2 酪氨酸磷酸酶的缺失导致与年龄相关的视网膜退化。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41419-024-06924-y
Ammaji Rajala, Rahul Rajala, Mohd A Bhat, Mark Eminhizer, Jeff Hao, Jianhai Du, Raju V S Rajala

Shp2, a critical SH2-domain-containing tyrosine phosphatase, is essential for cellular regulation and implicated in metabolic disruptions, obesity, diabetes, Noonan syndrome, LEOPARD syndrome, and cancers. This study focuses on Shp2 in rod photoreceptor cells, revealing its enrichment, particularly in rods. Deletion of Shp2 in rods leads to age-dependent photoreceptor degeneration. Shp2 targets occludin (OCLN), a tight junction protein, and its deletion reduces OCLN expression in the retina and retinal pigment epithelium (RPE). The isolation of actively translating mRNAs from rods lacking Shp2, followed by RNA sequencing, reveals alterations in cell cycle regulation. Additionally, altered retinal metabolism is observed in retinal cells lacking Shp2. Our studies indicate that Shp2 is crucial for maintaining the structure and function of photoreceptors.

Shp2是一种重要的含SH2域的酪氨酸磷酸酶,对细胞调控至关重要,与代谢紊乱、肥胖、糖尿病、努南综合征、LEOPARD综合征和癌症有关。本研究重点研究了杆状感光细胞中的 Shp2,揭示了其富集作用,尤其是在杆状细胞中。在杆状细胞中缺失 Shp2 会导致年龄依赖性光感受器退化。Shp2 的靶标是一种紧密连接蛋白--闭塞素(OCLN),其缺失会降低视网膜和视网膜色素上皮(RPE)中闭塞素的表达。从缺乏 Shp2 的视杆细胞中分离出活跃翻译的 mRNA,然后进行 RNA 测序,发现细胞周期调控发生了改变。此外,在缺乏 Shp2 的视网膜细胞中还观察到视网膜新陈代谢的改变。我们的研究表明,Shp2对维持感光细胞的结构和功能至关重要。
{"title":"Age-related retinal degeneration resulting from the deletion of Shp2 tyrosine phosphatase in photoreceptor neurons.","authors":"Ammaji Rajala, Rahul Rajala, Mohd A Bhat, Mark Eminhizer, Jeff Hao, Jianhai Du, Raju V S Rajala","doi":"10.1038/s41419-024-06924-y","DOIUrl":"10.1038/s41419-024-06924-y","url":null,"abstract":"<p><p>Shp2, a critical SH2-domain-containing tyrosine phosphatase, is essential for cellular regulation and implicated in metabolic disruptions, obesity, diabetes, Noonan syndrome, LEOPARD syndrome, and cancers. This study focuses on Shp2 in rod photoreceptor cells, revealing its enrichment, particularly in rods. Deletion of Shp2 in rods leads to age-dependent photoreceptor degeneration. Shp2 targets occludin (OCLN), a tight junction protein, and its deletion reduces OCLN expression in the retina and retinal pigment epithelium (RPE). The isolation of actively translating mRNAs from rods lacking Shp2, followed by RNA sequencing, reveals alterations in cell cycle regulation. Additionally, altered retinal metabolism is observed in retinal cells lacking Shp2. Our studies indicate that Shp2 is crucial for maintaining the structure and function of photoreceptors.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310310/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906052","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
Elucidating the role of S100A10 in CD8+ T cell exhaustion and HCC immune escape via the cPLA2 and 5-LOX axis. 通过 cPLA2 和 5-LOX 轴阐明 S100A10 在 CD8+ T 细胞衰竭和 HCC 免疫逃逸中的作用。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41419-024-06895-0
Ganggang Wang, Xiaowei Shen, Wenzhi Jin, Chao Song, Meiyuan Dong, Zhijie Zhou, Xiaoliang Wang

Hepatocellular carcinoma (HCC) is a common malignant tumor with a complex immune evasion mechanism posing a challenge to treatment. The role of the S100A10 gene in various cancers has garnered significant attention. This study aims to elucidate the impact of S100A10 on CD8+ T cell exhaustion via the cPLA2 and 5-LOX axis, thereby elucidating its role in immune evasion in HCC. By analyzing the HCC-related data from the GEO and TCGA databases, we identified differentially expressed genes associated with lipid metabolism and developed a prognostic risk model. Subsequently, through RNA-seq and PPI analyses, we determined vital lipid metabolism genes and downstream factors S100A10, ACOT7, and SMS, which were significantly correlated with CD8+ T cell infiltration. Given the most significant expression differences, we selected S100A10 for further investigation. Both in vitro and in vivo experiments were conducted, including co-culture experiments of CD8+ T cells with MHCC97-L cells, Co-IP experiments, and validation in an HCC mouse model. S100A10 was significantly overexpressed in HCC tissues and potentially regulates CD8+ T cell exhaustion and lipid metabolism reprogramming through the cPLA2 and 5-LOX axis. Silencing S100A10 could inhibit CD8+ T cell exhaustion, further suppressing immune evasion in HCC. S100A10 may activate the cPLA2 and 5-LOX axis, initiating lipid metabolism reprogramming and upregulating LTB4 levels, thus promoting CD8+ T cell exhaustion in HCC tissues, facilitating immune evasion by HCC cells, ultimately impacting the growth and migration of HCC cells. This research highlights the critical role of S100A10 via the cPLA2 and 5-LOX axis in immune evasion in HCC, providing new theoretical foundations and potential targets for diagnosing and treating HCC.

肝细胞癌(HCC)是一种常见的恶性肿瘤,其复杂的免疫逃避机制给治疗带来了挑战。S100A10 基因在各种癌症中的作用引起了广泛关注。本研究旨在阐明 S100A10 通过 cPLA2 和 5-LOX 轴对 CD8+ T 细胞衰竭的影响,从而阐明其在 HCC 免疫逃避中的作用。通过分析GEO和TCGA数据库中的HCC相关数据,我们发现了与脂质代谢相关的差异表达基因,并建立了预后风险模型。随后,通过 RNA-seq 和 PPI 分析,我们确定了与 CD8+ T 细胞浸润显著相关的重要脂质代谢基因和下游因子 S100A10、ACOT7 和 SMS。鉴于表达差异最为明显,我们选择了 S100A10 进行进一步研究。我们进行了体外和体内实验,包括 CD8+ T 细胞与 MHCC97-L 细胞的共培养实验、Co-IP 实验以及在 HCC 小鼠模型中的验证。S100A10在HCC组织中明显过表达,可能通过cPLA2和5-LOX轴调节CD8+ T细胞衰竭和脂质代谢重编程。沉默S100A10可抑制CD8+ T细胞衰竭,进一步抑制HCC的免疫逃避。S100A10可能会激活cPLA2和5-LOX轴,启动脂质代谢重编程并上调LTB4水平,从而促进HCC组织中CD8+ T细胞衰竭,促进HCC细胞的免疫逃避,最终影响HCC细胞的生长和迁移。这项研究强调了S100A10通过cPLA2和5-LOX轴在HCC免疫逃避中的关键作用,为诊断和治疗HCC提供了新的理论基础和潜在靶点。
{"title":"Elucidating the role of S100A10 in CD8<sup>+</sup> T cell exhaustion and HCC immune escape via the cPLA2 and 5-LOX axis.","authors":"Ganggang Wang, Xiaowei Shen, Wenzhi Jin, Chao Song, Meiyuan Dong, Zhijie Zhou, Xiaoliang Wang","doi":"10.1038/s41419-024-06895-0","DOIUrl":"10.1038/s41419-024-06895-0","url":null,"abstract":"<p><p>Hepatocellular carcinoma (HCC) is a common malignant tumor with a complex immune evasion mechanism posing a challenge to treatment. The role of the S100A10 gene in various cancers has garnered significant attention. This study aims to elucidate the impact of S100A10 on CD8<sup>+</sup> T cell exhaustion via the cPLA2 and 5-LOX axis, thereby elucidating its role in immune evasion in HCC. By analyzing the HCC-related data from the GEO and TCGA databases, we identified differentially expressed genes associated with lipid metabolism and developed a prognostic risk model. Subsequently, through RNA-seq and PPI analyses, we determined vital lipid metabolism genes and downstream factors S100A10, ACOT7, and SMS, which were significantly correlated with CD8<sup>+</sup> T cell infiltration. Given the most significant expression differences, we selected S100A10 for further investigation. Both in vitro and in vivo experiments were conducted, including co-culture experiments of CD8<sup>+</sup> T cells with MHCC97-L cells, Co-IP experiments, and validation in an HCC mouse model. S100A10 was significantly overexpressed in HCC tissues and potentially regulates CD8<sup>+</sup> T cell exhaustion and lipid metabolism reprogramming through the cPLA2 and 5-LOX axis. Silencing S100A10 could inhibit CD8<sup>+</sup> T cell exhaustion, further suppressing immune evasion in HCC. S100A10 may activate the cPLA2 and 5-LOX axis, initiating lipid metabolism reprogramming and upregulating LTB4 levels, thus promoting CD8<sup>+</sup> T cell exhaustion in HCC tissues, facilitating immune evasion by HCC cells, ultimately impacting the growth and migration of HCC cells. This research highlights the critical role of S100A10 via the cPLA2 and 5-LOX axis in immune evasion in HCC, providing new theoretical foundations and potential targets for diagnosing and treating HCC.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310305/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906016","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
Fetal growth restriction induced by maternal gal-3 deficiency is associated with altered gut-placenta axis. 母体缺乏 gal-3 导致的胎儿生长受限与肠道-胎盘轴的改变有关。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41419-024-06962-6
Yiran Xie, Fangqi Zhao, Yiru Wang, Sophia Borowski, Nancy Freitag, Irene Tirado-Gonzalez, Naomi Hofsink, Urte Matschl, Torsten Plösch, Mariana G Garcia, Sandra M Blois

Adverse intrauterine conditions may cause fetal growth restriction (FGR), a pregnancy complication frequently linked to perinatal morbidity and mortality. Although many studies have focused on FGR, the pathophysiological processes underlying this disorder are complex and incompletely understood. We have recently determined that galectin-3 (gal-3), a β-galactoside-binding protein, regulates pregnancy-associated processes, including uterine receptibility, maternal vascular adaptation and placentation. Because gal-3 is expressed at both sides of the maternal-fetal interface, we unraveled the contribution of maternal- and paternal-derived gal-3 on fetal-placental development in the prenatal window and its effects on the post-natal period. Deficiency of maternal gal-3 induced maternal gut microbiome dysbiosis, resulting in a sex-specific fetal growth restriction mainly observed in female fetuses and offspring. In addition, poor placental metabolic adaptions (characterized by decreased trophoblast glycogen content and insulin-like growth factor 2 (Igf2) gene hypomethylation) were only associated with a lack of maternal-derived gal-3. Paternal gal-3 deficiency caused compromised vascularization in the placental labyrinth without affecting fetal growth trajectory. Thus, maternal-derived gal-3 may play a key role in fetal-placental development through the gut-placenta axis.

不利的宫内条件可能会导致胎儿生长受限(FGR),这是一种妊娠并发症,经常与围产期发病率和死亡率有关。尽管许多研究都聚焦于 FGR,但这种疾病的病理生理过程却十分复杂,人们对其了解也不全面。我们最近发现,半乳糖苷结合蛋白 galectin-3(gal-3)可调节妊娠相关过程,包括子宫接受性、母体血管适应性和胎盘形成。由于gal-3在母胎界面两侧均有表达,我们揭示了母源和父源gal-3在产前窗口期对胎儿-胎盘发育的贡献及其对产后的影响。母源gal-3的缺乏会诱发母体肠道微生物群失调,导致胎儿性别特异性生长受限,这种情况主要出现在雌性胎儿和后代身上。此外,胎盘代谢适应不良(表现为滋养层糖原含量降低和胰岛素样生长因子2(Igf2)基因低甲基化)仅与母源gal-3缺乏有关。父源gal-3缺乏会导致胎盘迷宫血管化受损,但不会影响胎儿的生长轨迹。因此,母源gal-3可能通过肠道-胎盘轴在胎儿-胎盘发育过程中发挥关键作用。
{"title":"Fetal growth restriction induced by maternal gal-3 deficiency is associated with altered gut-placenta axis.","authors":"Yiran Xie, Fangqi Zhao, Yiru Wang, Sophia Borowski, Nancy Freitag, Irene Tirado-Gonzalez, Naomi Hofsink, Urte Matschl, Torsten Plösch, Mariana G Garcia, Sandra M Blois","doi":"10.1038/s41419-024-06962-6","DOIUrl":"10.1038/s41419-024-06962-6","url":null,"abstract":"<p><p>Adverse intrauterine conditions may cause fetal growth restriction (FGR), a pregnancy complication frequently linked to perinatal morbidity and mortality. Although many studies have focused on FGR, the pathophysiological processes underlying this disorder are complex and incompletely understood. We have recently determined that galectin-3 (gal-3), a β-galactoside-binding protein, regulates pregnancy-associated processes, including uterine receptibility, maternal vascular adaptation and placentation. Because gal-3 is expressed at both sides of the maternal-fetal interface, we unraveled the contribution of maternal- and paternal-derived gal-3 on fetal-placental development in the prenatal window and its effects on the post-natal period. Deficiency of maternal gal-3 induced maternal gut microbiome dysbiosis, resulting in a sex-specific fetal growth restriction mainly observed in female fetuses and offspring. In addition, poor placental metabolic adaptions (characterized by decreased trophoblast glycogen content and insulin-like growth factor 2 (Igf2) gene hypomethylation) were only associated with a lack of maternal-derived gal-3. Paternal gal-3 deficiency caused compromised vascularization in the placental labyrinth without affecting fetal growth trajectory. Thus, maternal-derived gal-3 may play a key role in fetal-placental development through the gut-placenta axis.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310209/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906017","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
Preservation of retinal structure and function in two mouse models of inherited retinal degeneration by ONL1204, an inhibitor of the Fas receptor. 通过 Fas 受体抑制剂 ONL1204 保护两种遗传性视网膜变性小鼠模型的视网膜结构和功能。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-08 DOI: 10.1038/s41419-024-06970-6
Mengling Yang, Jingyu Yao, Lin Jia, Andrew J Kocab, David N Zacks

Due to the large number of genes and mutations that result in inherited retinal degenerations (IRD), there has been a paucity of therapeutic options for these patients. There is a large unmet need for therapeutic approaches targeting shared pathophysiologic pathways in a mutation-independent manner. The Fas receptor is a major activator and regulator of retinal cell death and inflammation in a variety of ocular diseases. We previously reported the activation of Fas-mediated photoreceptor (PR) cell death in two different IRD mouse models, rd10 and P23H, and demonstrated the protective effect of genetic Fas inhibition. The purpose of this study was to examine the effects of pharmacologic inhibition of Fas in these two models by intravitreal injection with a small peptide inhibitor of the Fas receptor, ONL1204. A single intravitreal injection of ONL1204 was given to one eye of rd10 mice at P14. Two intravitreal injections of ONL1204 were given to the P23H mice, once at P14 and again at 2-months of age. The fellow eyes were injected with vehicle alone. Fas activation, rate of PR cell death, retinal function, and the activation of immune cells in the retina were evaluated. In both rd10 and P23H mice, ONL1204 treatment resulted in decreased number of TUNEL (+) PRs, decreased caspase 8 activity, enhanced photoreceptor cell counts, and improved visual function compared with vehicle treated fellow eyes. Treatment with ONL1204 also reduced immune cell activation in the retinas of both rd10 and P23H mice. The protective effect of pharmacologic inhibition of Fas by ONL1204 in two distinct mouse models of retinal degeneration suggests that targeting this common pathophysiologic mechanism of cell death and inflammation represents a potential therapeutic approach to preserve the retina in patients with IRD, regardless of the genetic underpinning.

由于导致遗传性视网膜变性(IRD)的基因和突变数量众多,针对这些患者的治疗方案一直很少。以不依赖基因突变的方式针对共同病理生理途径的治疗方法还有大量需求没有得到满足。Fas 受体是多种眼部疾病中视网膜细胞死亡和炎症的主要激活剂和调节剂。我们以前曾报道过在两种不同的 IRD 小鼠模型(rd10 和 P23H)中 Fas 介导的感光细胞(PR)死亡的激活,并证明了遗传性 Fas 抑制的保护作用。本研究的目的是通过玻璃体内注射 Fas 受体的小肽抑制剂 ONL1204 来检测药理学抑制 Fas 对这两种模型的影响。rd10小鼠的一只眼睛在P14时接受了一次ONL1204的玻璃体内注射。对P23H小鼠进行了两次ONL1204玻璃体内注射,一次是在P14岁时,另一次是在2月龄时。同组小鼠的眼睛只注射了药物。对 Fas 激活、PR 细胞死亡率、视网膜功能和视网膜中免疫细胞的激活情况进行了评估。在rd10和P23H小鼠中,ONL1204治疗导致TUNEL(+)PR细胞数量减少,caspase 8活性降低,感光细胞数量增加,视觉功能比用药物治疗的同组小鼠的眼睛有所改善。用 ONL1204 治疗还能减少 rd10 和 P23H 小鼠视网膜中免疫细胞的活化。在两种不同的视网膜变性小鼠模型中,ONL1204对Fas的药理抑制具有保护作用,这表明针对细胞死亡和炎症的这一共同病理生理机制是保护IRD患者视网膜的一种潜在治疗方法,无论其遗传基础如何。
{"title":"Preservation of retinal structure and function in two mouse models of inherited retinal degeneration by ONL1204, an inhibitor of the Fas receptor.","authors":"Mengling Yang, Jingyu Yao, Lin Jia, Andrew J Kocab, David N Zacks","doi":"10.1038/s41419-024-06970-6","DOIUrl":"10.1038/s41419-024-06970-6","url":null,"abstract":"<p><p>Due to the large number of genes and mutations that result in inherited retinal degenerations (IRD), there has been a paucity of therapeutic options for these patients. There is a large unmet need for therapeutic approaches targeting shared pathophysiologic pathways in a mutation-independent manner. The Fas receptor is a major activator and regulator of retinal cell death and inflammation in a variety of ocular diseases. We previously reported the activation of Fas-mediated photoreceptor (PR) cell death in two different IRD mouse models, rd10 and P23H, and demonstrated the protective effect of genetic Fas inhibition. The purpose of this study was to examine the effects of pharmacologic inhibition of Fas in these two models by intravitreal injection with a small peptide inhibitor of the Fas receptor, ONL1204. A single intravitreal injection of ONL1204 was given to one eye of rd10 mice at P14. Two intravitreal injections of ONL1204 were given to the P23H mice, once at P14 and again at 2-months of age. The fellow eyes were injected with vehicle alone. Fas activation, rate of PR cell death, retinal function, and the activation of immune cells in the retina were evaluated. In both rd10 and P23H mice, ONL1204 treatment resulted in decreased number of TUNEL (+) PRs, decreased caspase 8 activity, enhanced photoreceptor cell counts, and improved visual function compared with vehicle treated fellow eyes. Treatment with ONL1204 also reduced immune cell activation in the retinas of both rd10 and P23H mice. The protective effect of pharmacologic inhibition of Fas by ONL1204 in two distinct mouse models of retinal degeneration suggests that targeting this common pathophysiologic mechanism of cell death and inflammation represents a potential therapeutic approach to preserve the retina in patients with IRD, regardless of the genetic underpinning.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11310419/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141906019","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
ALDH1A1 promotes immune escape of tumor cells through ZBTB7B-glycolysis pathway. ALDH1A1通过ZBTB7B-糖酵解途径促进肿瘤细胞的免疫逃逸。
IF 8.1 1区 生物学 Q1 CELL BIOLOGY Pub Date : 2024-08-07 DOI: 10.1038/s41419-024-06943-9
Mingyuan Wang, Taoli Wang, Jinjin Wang, Yuexin Yang, Xi Li, Huan Chen, Jingnan Liao

The primary impediment to the success of immunotherapy lies in the immune evasion orchestrated by tumors, contributing to the suboptimal overall response rates observed. Despite this recognition, the intricacies of the underlying mechanisms remain incompletely understood. Through preliminary detection of clinical patient tissues, we have found that ALDH1A1 was a key gene for the prognosis of cancer patients and tumor glycolysis. In vitro experiments and tumor formation in nude mice suggested that targeting ALDH1A1 could inhibit tumor growth. Through further analysis of xenograft tumor models in immune-normal mice and flow cytometry, we found that deficiency in ALDH1A1 could promote immune system suppression of tumors in vivo. Specifically, RNA-seq analysis, combined with qPCR and western blot, identified the transcription factor ZBTB7B as downstream of ALDH1A1. The binding sites of the transcription factor ZBTB7B on the LDHA promoter region, which is responsible for regulating the rate-limiting enzyme gene LDHA in glycolysis, were determined using luciferase reporter gene detection and Chip-qPCR, respectively. In addition, the increased SUMOylation of ZBTB7B stabilized its transcriptional activity. Further in vivo and in vitro experiments confirmed that the combination of targeting ALDH1A1 and ZBTB7B with immune checkpoint inhibitors could synergistically inhibit tumors in vivo. Finally, after conducting additional verification of patient tissue and clinical data, we have confirmed the potential translational value of targeting ALDH1A1 and ZBTB7B for tumor immunotherapy. These results emphasize the potential translational significance of targeting ALDH1A1 and ZBTB7B in the realm of tumor immunotherapy. The convergence of ALDH1A1 inhibition and immune checkpoint blockade, particularly with PD-L1/PD-1 mAb, presents a compelling avenue for curtailing tumor immune escape.

免疫疗法取得成功的主要障碍在于肿瘤精心策划的免疫逃避,这也是导致总体反应率不理想的原因之一。尽管人们认识到了这一点,但对其潜在机制的复杂性仍不甚了解。通过对临床患者组织的初步检测,我们发现 ALDH1A1 是影响癌症患者预后和肿瘤糖酵解的关键基因。体外实验和裸鼠肿瘤形成表明,靶向 ALDH1A1 可抑制肿瘤生长。通过进一步分析免疫正常小鼠的异种移植肿瘤模型和流式细胞术,我们发现 ALDH1A1 的缺乏可促进体内免疫系统对肿瘤的抑制。具体来说,RNA-seq分析结合qPCR和Western blot,确定了转录因子ZBTB7B是ALDH1A1的下游。利用荧光素酶报告基因检测和芯片-qPCR,分别确定了转录因子ZBTB7B在LDHA启动子区域的结合位点,而LDHA启动子区域负责调控糖酵解过程中的限速酶基因LDHA。此外,ZBTB7B SUMOylation 的增加稳定了其转录活性。进一步的体内和体外实验证实,靶向 ALDH1A1 和 ZBTB7B 与免疫检查点抑制剂联合使用可协同抑制体内肿瘤。最后,经过对患者组织和临床数据的进一步验证,我们证实了靶向 ALDH1A1 和 ZBTB7B 用于肿瘤免疫治疗的潜在转化价值。这些结果强调了靶向 ALDH1A1 和 ZBTB7B 在肿瘤免疫治疗领域的潜在转化意义。ALDH1A1抑制与免疫检查点阻断(尤其是PD-L1/PD-1 mAb)的结合为遏制肿瘤免疫逃逸提供了一条令人信服的途径。
{"title":"ALDH1A1 promotes immune escape of tumor cells through ZBTB7B-glycolysis pathway.","authors":"Mingyuan Wang, Taoli Wang, Jinjin Wang, Yuexin Yang, Xi Li, Huan Chen, Jingnan Liao","doi":"10.1038/s41419-024-06943-9","DOIUrl":"10.1038/s41419-024-06943-9","url":null,"abstract":"<p><p>The primary impediment to the success of immunotherapy lies in the immune evasion orchestrated by tumors, contributing to the suboptimal overall response rates observed. Despite this recognition, the intricacies of the underlying mechanisms remain incompletely understood. Through preliminary detection of clinical patient tissues, we have found that ALDH1A1 was a key gene for the prognosis of cancer patients and tumor glycolysis. In vitro experiments and tumor formation in nude mice suggested that targeting ALDH1A1 could inhibit tumor growth. Through further analysis of xenograft tumor models in immune-normal mice and flow cytometry, we found that deficiency in ALDH1A1 could promote immune system suppression of tumors in vivo. Specifically, RNA-seq analysis, combined with qPCR and western blot, identified the transcription factor ZBTB7B as downstream of ALDH1A1. The binding sites of the transcription factor ZBTB7B on the LDHA promoter region, which is responsible for regulating the rate-limiting enzyme gene LDHA in glycolysis, were determined using luciferase reporter gene detection and Chip-qPCR, respectively. In addition, the increased SUMOylation of ZBTB7B stabilized its transcriptional activity. Further in vivo and in vitro experiments confirmed that the combination of targeting ALDH1A1 and ZBTB7B with immune checkpoint inhibitors could synergistically inhibit tumors in vivo. Finally, after conducting additional verification of patient tissue and clinical data, we have confirmed the potential translational value of targeting ALDH1A1 and ZBTB7B for tumor immunotherapy. These results emphasize the potential translational significance of targeting ALDH1A1 and ZBTB7B in the realm of tumor immunotherapy. The convergence of ALDH1A1 inhibition and immune checkpoint blockade, particularly with PD-L1/PD-1 mAb, presents a compelling avenue for curtailing tumor immune escape.</p>","PeriodicalId":9734,"journal":{"name":"Cell Death & Disease","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11303523/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141896903","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