首页 > 最新文献

Oncogenesis最新文献

英文 中文
An E3 ligase TRIM1 promotes colorectal cancer progression via K63-linked ubiquitination and activation of HIF1α. E3连接酶TRIM1通过与K63连接的泛素化和激活HIF1α促进结直肠癌的进展。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-20 DOI: 10.1038/s41389-024-00517-2
Liuliu Shi, Xianglan Fang, Lijie Du, Jin Yang, Juan Xue, Xiaokai Yue, Duoshuang Xie, Yuanjian Hui, Kun Meng

Accumulating studies have shown that E3 ligases play crucial roles in regulating cellular biological processes and signaling pathways during carcinogenesis via ubiquitination. Tripartite-motif (TRIM) ubiquitin E3 ligases consist of over 70 members. However, the clinical significance and their contributions to tumorigenesis remain largely unknown. In this study, we analyzed the RNA-sequencing expression of TRIM E3 ligases in colorectal cancer (CRC) and identified 10 differentially expressed genes, among which TRIM1 expression predicted poor prognosis of CRC patients. We demonstrated that TRIM1 expression is positively associated with CRC pathological stages, and higher expression is positively correlated with infiltrating levels of immune cells and immunotherapy biomarkers. TRIM1 expression promotes the proliferation and migration of colorectal cancer cells in vitro and in vivo. Transcriptional analysis showed that TRIM1 is responsible for metabolism promotion and immune suppression. Mechanistically, we found that TRIM1 binds HIF1α and mediates its K63-linked ubiquitination, which is required for HIF1α nuclear translocation and subsequent activation. Ubiquitination occurs at Lys214 in the loop between the two PAS domains of HIF1α, and mutation of Lys214 severely disturbs the function of HIF1α. Besides, HIF1α ubiquitination enhances its binding with proteins involved in cellular trafficking and nucleocytoplasmic transport pathway. Collectively, our results indicate TRIM1's role in predicting prognosis and reveal how TRIM1 functions to upregulate HIF1α expression and promote tumor cell proliferation.

越来越多的研究表明,E3 连接酶通过泛素化作用在致癌过程中调节细胞生物过程和信号通路方面发挥着至关重要的作用。三态泛素 E3 连接酶(TRIM)由 70 多个成员组成。然而,它们的临床意义及其对肿瘤发生的贡献在很大程度上仍不为人所知。在这项研究中,我们分析了 TRIM E3 连接酶在结直肠癌(CRC)中的 RNA 序列表达,并确定了 10 个差异表达基因,其中 TRIM1 的表达预示着 CRC 患者的不良预后。我们证实,TRIM1的表达与CRC的病理分期呈正相关,较高的表达与免疫细胞的浸润水平和免疫疗法生物标志物呈正相关。TRIM1 的表达促进了结直肠癌细胞在体外和体内的增殖和迁移。转录分析表明,TRIM1 负责促进新陈代谢和抑制免疫。从机理上讲,我们发现 TRIM1 与 HIF1α 结合并介导其 K63 链接泛素化,这是 HIF1α 核转位和随后激活所必需的。泛素化发生在 HIF1α 两个 PAS 结构域之间环路的 Lys214 上,Lys214 的突变会严重干扰 HIF1α 的功能。此外,HIF1α泛素化会增强其与参与细胞运输和核胞质运输途径的蛋白的结合。总之,我们的研究结果表明了TRIM1在预测预后中的作用,并揭示了TRIM1是如何上调HIF1α表达和促进肿瘤细胞增殖的。
{"title":"An E3 ligase TRIM1 promotes colorectal cancer progression via K63-linked ubiquitination and activation of HIF1α.","authors":"Liuliu Shi, Xianglan Fang, Lijie Du, Jin Yang, Juan Xue, Xiaokai Yue, Duoshuang Xie, Yuanjian Hui, Kun Meng","doi":"10.1038/s41389-024-00517-2","DOIUrl":"10.1038/s41389-024-00517-2","url":null,"abstract":"<p><p>Accumulating studies have shown that E3 ligases play crucial roles in regulating cellular biological processes and signaling pathways during carcinogenesis via ubiquitination. Tripartite-motif (TRIM) ubiquitin E3 ligases consist of over 70 members. However, the clinical significance and their contributions to tumorigenesis remain largely unknown. In this study, we analyzed the RNA-sequencing expression of TRIM E3 ligases in colorectal cancer (CRC) and identified 10 differentially expressed genes, among which TRIM1 expression predicted poor prognosis of CRC patients. We demonstrated that TRIM1 expression is positively associated with CRC pathological stages, and higher expression is positively correlated with infiltrating levels of immune cells and immunotherapy biomarkers. TRIM1 expression promotes the proliferation and migration of colorectal cancer cells in vitro and in vivo. Transcriptional analysis showed that TRIM1 is responsible for metabolism promotion and immune suppression. Mechanistically, we found that TRIM1 binds HIF1α and mediates its K63-linked ubiquitination, which is required for HIF1α nuclear translocation and subsequent activation. Ubiquitination occurs at Lys214 in the loop between the two PAS domains of HIF1α, and mutation of Lys214 severely disturbs the function of HIF1α. Besides, HIF1α ubiquitination enhances its binding with proteins involved in cellular trafficking and nucleocytoplasmic transport pathway. Collectively, our results indicate TRIM1's role in predicting prognosis and reveal how TRIM1 functions to upregulate HIF1α expression and promote tumor cell proliferation.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11106307/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141071647","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NAC1 transcriptional activation of LDHA induces hepatitis B virus immune evasion leading to cirrhosis and hepatocellular carcinoma development NAC1 对 LDHA 的转录激活诱导乙型肝炎病毒免疫逃避,导致肝硬化和肝细胞癌的发展
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-05-04 DOI: 10.1038/s41389-024-00515-4
Wenbiao Chen, Liliangzi Guo, Huixuan Xu, Yong Dai, Jun Yao, Lisheng Wang

Our study aimed to elucidate the molecular mechanisms underlying NAC1 (nucleus accumbens associated 1) transcriptional regulation of LDHA and its role in HBV immune evasion, thus contributing to the development of cirrhosis and hepatocellular carcinoma (HCC). Utilizing public datasets, we performed differential gene expression and weighted gene co-expression network analysis (WGCNA) on HBV-induced cirrhosis/HCC data. We identified candidate genes by intersecting differentially expressed genes with co-expression modules. We validated these genes using the TCGA database, conducting survival analysis to pinpoint key genes affecting HBV-HCC prognosis. We also employed the TIMER database for immune cell infiltration data and analyzed correlations with identified key genes to uncover potential immune escape pathways. In vitro, we investigated the impact of NAC1 and LDHA on immune cell apoptosis and HBV immune evasion. In vivo, we confirmed these findings using an HBV-induced cirrhosis model. Bioinformatics analysis revealed 676 genes influenced by HBV infection, with 475 genes showing differential expression in HBV-HCC. NAC1 emerged as a key gene, potentially mediating HBV immune escape through LDHA transcriptional regulation. Experimental data demonstrated that NAC1 transcriptionally activates LDHA, promoting immune cell apoptosis and HBV immune evasion. Animal studies confirmed these findings, linking NAC1-mediated LDHA activation to cirrhosis and HCC development. NAC1, highly expressed in HBV-infected liver cells, likely drives HBV immune escape by activating LDHA expression, inhibiting CD8 + T cells, and promoting cirrhosis and HCC development.

我们的研究旨在阐明NAC1(nucleus accumbens associated 1)转录调控LDHA的分子机制及其在HBV免疫逃避中的作用,从而促进肝硬化和肝细胞癌(HCC)的发展。利用公开数据集,我们对 HBV 诱导的肝硬化/肝癌数据进行了差异基因表达和加权基因共表达网络分析(WGCNA)。我们通过差异表达基因与共表达模块的交叉来确定候选基因。我们利用 TCGA 数据库验证了这些基因,并进行了生存分析,以确定影响 HBV-HCC 预后的关键基因。我们还利用 TIMER 数据库获取了免疫细胞浸润数据,并分析了与已确定关键基因的相关性,从而发现了潜在的免疫逃逸途径。在体外,我们研究了 NAC1 和 LDHA 对免疫细胞凋亡和 HBV 免疫逃避的影响。在体内,我们利用 HBV 诱导的肝硬化模型证实了这些发现。生物信息学分析揭示了 676 个受 HBV 感染影响的基因,其中 475 个基因在 HBV-HCC 中表现出差异表达。NAC1是一个关键基因,可能通过LDHA转录调控介导HBV免疫逃逸。实验数据表明,NAC1 可转录激活 LDHA,促进免疫细胞凋亡和 HBV 免疫逃避。动物实验证实了这些发现,并将 NAC1 介导的 LDHA 激活与肝硬化和 HCC 的发展联系起来。NAC1 在受 HBV 感染的肝细胞中高度表达,可能通过激活 LDHA 表达、抑制 CD8 + T 细胞、促进肝硬化和 HCC 的发展来推动 HBV 免疫逃避。
{"title":"NAC1 transcriptional activation of LDHA induces hepatitis B virus immune evasion leading to cirrhosis and hepatocellular carcinoma development","authors":"Wenbiao Chen, Liliangzi Guo, Huixuan Xu, Yong Dai, Jun Yao, Lisheng Wang","doi":"10.1038/s41389-024-00515-4","DOIUrl":"https://doi.org/10.1038/s41389-024-00515-4","url":null,"abstract":"<p>Our study aimed to elucidate the molecular mechanisms underlying NAC1 (nucleus accumbens associated 1) transcriptional regulation of LDHA and its role in HBV immune evasion, thus contributing to the development of cirrhosis and hepatocellular carcinoma (HCC). Utilizing public datasets, we performed differential gene expression and weighted gene co-expression network analysis (WGCNA) on HBV-induced cirrhosis/HCC data. We identified candidate genes by intersecting differentially expressed genes with co-expression modules. We validated these genes using the TCGA database, conducting survival analysis to pinpoint key genes affecting HBV-HCC prognosis. We also employed the TIMER database for immune cell infiltration data and analyzed correlations with identified key genes to uncover potential immune escape pathways. In vitro, we investigated the impact of NAC1 and LDHA on immune cell apoptosis and HBV immune evasion. In vivo, we confirmed these findings using an HBV-induced cirrhosis model. Bioinformatics analysis revealed 676 genes influenced by HBV infection, with 475 genes showing differential expression in HBV-HCC. NAC1 emerged as a key gene, potentially mediating HBV immune escape through LDHA transcriptional regulation. Experimental data demonstrated that NAC1 transcriptionally activates LDHA, promoting immune cell apoptosis and HBV immune evasion. Animal studies confirmed these findings, linking NAC1-mediated LDHA activation to cirrhosis and HCC development. NAC1, highly expressed in HBV-infected liver cells, likely drives HBV immune escape by activating LDHA expression, inhibiting CD8 + T cells, and promoting cirrhosis and HCC development.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-05-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140826988","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phenotyping of cancer-associated somatic mutations in the BCL2 transmembrane domain BCL2跨膜结构域癌症相关体细胞突变的表型分析
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-04-26 DOI: 10.1038/s41389-024-00516-3
Diego Leiva, Estefanía Lucendo, Alicia Belén García-Jareño, Mónica Sancho, Mar Orzáez

The BCL2 family of proteins controls cell death by modulating the permeabilization of the mitochondrial outer membrane through a fine-tuned equilibrium of interactions among anti- and pro-apoptotic members. The upregulation of anti-apoptotic BCL2 proteins represents an unfavorable prognostic factor in many tumor types due to their ability to shift the equilibrium toward cancer cell survival. Furthermore, cancer-associated somatic mutations in BCL2 genes interfere with the protein interaction network, thereby promoting cell survival. A range of studies have documented how these mutations affect the interactions between the cytosolic domains of BCL2 and evaluate the impact on cell death; however, as the BCL2 transmembrane interaction network remains poorly understood, somatic mutations affecting transmembrane regions have been classified as pathogenic-based solely on prediction algorithms. We comprehensively investigated cancer-associated somatic mutations affecting the transmembrane domain of BCL2 proteins and elucidated their effect on membrane insertion, hetero-interactions with the pro-apoptotic protein BAX, and modulation of cell death in cancer cells. Our findings reveal how specific mutations disrupt switchable interactions, alter the modulation of apoptosis, and contribute to cancer cell survival. These results provide experimental evidence to distinguish BCL2 transmembrane driver mutations from passenger mutations and provide new insight regarding selecting precision anti-tumor treatments.

BCL2 蛋白家族通过微调抗凋亡和促凋亡成员之间的相互作用平衡,调节线粒体外膜的通透性,从而控制细胞死亡。在许多肿瘤类型中,抗凋亡 BCL2 蛋白的上调是一个不利的预后因素,因为它们能够使平衡转向癌细胞的存活。此外,与癌症相关的 BCL2 基因体细胞突变会干扰蛋白质相互作用网络,从而促进细胞存活。一系列研究记录了这些突变如何影响BCL2细胞膜结构域之间的相互作用,并评估了其对细胞死亡的影响;然而,由于人们对BCL2跨膜相互作用网络仍然知之甚少,影响跨膜区域的体细胞突变被归类为致病性突变完全是基于预测算法。我们全面研究了影响BCL2蛋白跨膜结构域的癌症相关体细胞突变,阐明了它们对癌细胞膜插入、与促凋亡蛋白BAX的异质相互作用以及细胞死亡调控的影响。我们的研究结果揭示了特定突变是如何破坏可切换的相互作用、改变对细胞凋亡的调控以及促进癌细胞存活的。这些结果为区分BCL2跨膜驱动突变和乘客突变提供了实验证据,并为选择精确的抗肿瘤治疗方法提供了新的见解。
{"title":"Phenotyping of cancer-associated somatic mutations in the BCL2 transmembrane domain","authors":"Diego Leiva, Estefanía Lucendo, Alicia Belén García-Jareño, Mónica Sancho, Mar Orzáez","doi":"10.1038/s41389-024-00516-3","DOIUrl":"https://doi.org/10.1038/s41389-024-00516-3","url":null,"abstract":"<p>The BCL2 family of proteins controls cell death by modulating the permeabilization of the mitochondrial outer membrane through a fine-tuned equilibrium of interactions among anti- and pro-apoptotic members. The upregulation of anti-apoptotic BCL2 proteins represents an unfavorable prognostic factor in many tumor types due to their ability to shift the equilibrium toward cancer cell survival. Furthermore, cancer-associated somatic mutations in <i>BCL2</i> genes interfere with the protein interaction network, thereby promoting cell survival. A range of studies have documented how these mutations affect the interactions between the cytosolic domains of BCL2 and evaluate the impact on cell death; however, as the BCL2 transmembrane interaction network remains poorly understood, somatic mutations affecting transmembrane regions have been classified as pathogenic-based solely on prediction algorithms. We comprehensively investigated cancer-associated somatic mutations affecting the transmembrane domain of BCL2 proteins and elucidated their effect on membrane insertion, hetero-interactions with the pro-apoptotic protein BAX, and modulation of cell death in cancer cells. Our findings reveal how specific mutations disrupt switchable interactions, alter the modulation of apoptosis, and contribute to cancer cell survival. These results provide experimental evidence to distinguish BCL2 transmembrane driver mutations from passenger mutations and provide new insight regarding selecting precision anti-tumor treatments.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140803172","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Acidovorax temperans skews neutrophil maturation and polarizes Th17 cells to promote lung adenocarcinoma development 嗜酸性粒细胞扭曲了中性粒细胞的成熟并使Th17细胞极化,从而促进了肺腺癌的发展
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-04-03 DOI: 10.1038/s41389-024-00513-6
Joshua K. Stone, Natalia von Muhlinen, Chenran Zhang, Ana I. Robles, Amy L. Flis, Eleazar Vega-Valle, Akihiko Miyanaga, Masaru Matsumoto, K. Leigh Greathouse, Tomer Cooks, Giorgio Trinchieri, Curtis C. Harris

Change within the intratumoral microbiome is a common feature in lung and other cancers and may influence inflammation and immunity in the tumor microenvironment, affecting growth and metastases. We previously characterized the lung cancer microbiome in patients and identified Acidovorax temperans as enriched in tumors. Here, we instilled A. temperans in an animal model driven by mutant K-ras and Tp53. This revealed A. temperans accelerates tumor development and burden through infiltration of proinflammatory cells. Neutrophils exposed to A. temperans displayed a mature, pro-tumorigenic phenotype with increased cytokine signaling, with a global shift away from IL-1β signaling. Neutrophil to monocyte and macrophage signaling upregulated MHC II to activate CD4+ T cells, polarizing them to an IL-17A+ phenotype detectable in CD4+ and γδ populations (T17). These T17 cells shared a common gene expression program predictive of poor survival in human LUAD. These data indicate bacterial exposure promotes tumor growth by modulating inflammation.

瘤内微生物组的变化是肺癌和其他癌症的共同特征,可能会影响肿瘤微环境中的炎症和免疫,从而影响生长和转移。我们以前曾对患者的肺癌微生物组进行过鉴定,发现Acidovorax temperans富集在肿瘤中。在这里,我们在一个由突变 K-ras 和 Tp53 驱动的动物模型中灌输了节肢动物。这表明A. temperans通过促炎细胞的浸润加速了肿瘤的发展和负担。暴露于A. temperans的中性粒细胞表现出成熟的促肿瘤表型,细胞因子信号转导增加,IL-1β信号转导发生全面转移。中性粒细胞到单核细胞和巨噬细胞的信号传导上调了 MHC II,从而激活了 CD4+ T 细胞,使其极化为可在 CD4+ 和 γδ 群体(T17)中检测到的 IL-17A+ 表型。这些T17细胞具有共同的基因表达程序,可预测人类LUAD的不良存活率。这些数据表明,细菌暴露会通过调节炎症促进肿瘤生长。
{"title":"Acidovorax temperans skews neutrophil maturation and polarizes Th17 cells to promote lung adenocarcinoma development","authors":"Joshua K. Stone, Natalia von Muhlinen, Chenran Zhang, Ana I. Robles, Amy L. Flis, Eleazar Vega-Valle, Akihiko Miyanaga, Masaru Matsumoto, K. Leigh Greathouse, Tomer Cooks, Giorgio Trinchieri, Curtis C. Harris","doi":"10.1038/s41389-024-00513-6","DOIUrl":"https://doi.org/10.1038/s41389-024-00513-6","url":null,"abstract":"<p>Change within the intratumoral microbiome is a common feature in lung and other cancers and may influence inflammation and immunity in the tumor microenvironment, affecting growth and metastases. We previously characterized the lung cancer microbiome in patients and identified <i>Acidovorax temperans</i> as enriched in tumors. Here, we instilled <i>A. temperans</i> in an animal model driven by mutant K-ras and Tp53. This revealed <i>A. temperans</i> accelerates tumor development and burden through infiltration of proinflammatory cells. Neutrophils exposed to <i>A. temperans</i> displayed a mature, pro-tumorigenic phenotype with increased cytokine signaling, with a global shift away from IL-1β signaling. Neutrophil to monocyte and macrophage signaling upregulated MHC II to activate CD4<sup>+</sup> T cells, polarizing them to an IL-17A<sup>+</sup> phenotype detectable in CD4<sup>+</sup> and γδ populations (T17). These T17 cells shared a common gene expression program predictive of poor survival in human LUAD. These data indicate bacterial exposure promotes tumor growth by modulating inflammation.</p><figure></figure>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140600136","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
RIOK3 sustains colorectal cancer cell survival under glucose deprivation via an HSP90α-dependent pathway RIOK3 通过 HSP90α 依赖性途径维持葡萄糖剥夺条件下结直肠癌细胞的存活
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-07 DOI: 10.1038/s41389-024-00514-5
Nan Zhang, Lu Dong, Tingting Ning, Feng Du, Mengran Zhao, Junxuan Xu, Sian Xie, Si Liu, Xiujing Sun, Peng Li, Shutian Zhang, Shengtao Zhu

Glucose oxidation via the pentose phosphate pathway serves as the primary cellular mechanism for generating nicotinamide adenine dinucleotide phosphate (NADPH). The central regions of solid tumors typically experience glucose deficiency, emphasizing the need for sustained NADPH production crucial to tumor cell survival. This study highlights the crucial role of RIOK3 in maintaining NADPH production and colorectal cancer (CRC) cell survival during glucose deficiency. Our findings revealed upregulated RIOK3 expression upon glucose deprivation, with RIOK3 knockout significantly reducing cancer cell survival. Mechanistically, RIOK3 interacts with heat shock protein 90α (HSP90α), a chaperone integral to various cellular processes, thereby facilitating HSP90α binding to isocitrate dehydrogenase 1 (IDH1). This interaction further upregulates IDH1 expression, enhancing NADPH production and preserving redox balance. Furthermore, RIOK3 inhibition had no discernible effect on intracellular NADPH levels and cell death rates in HSP90α-knockdown cells. Collectively, our findings suggest that RIOK3 sustains colon cancer cell survival in low-glucose environments through an HSP90α-dependent pathway. This highlights the significance of the RIOK3–HSP90α–IDH1 cascade, providing insights into potential targeted therapeutic strategies for CRC in metabolic stress conditions.

通过磷酸戊糖途径进行的葡萄糖氧化是产生烟酰胺腺嘌呤二核苷酸磷酸(NADPH)的主要细胞机制。实体瘤的中心区域通常会出现葡萄糖缺乏,这就强调了持续产生 NADPH 的必要性,而 NADPH 对肿瘤细胞的存活至关重要。本研究强调了 RIOK3 在葡萄糖缺乏时维持 NADPH 生成和结直肠癌细胞存活的关键作用。我们的研究结果表明,在葡萄糖缺乏时,RIOK3表达上调,RIOK3敲除可显著降低癌细胞存活率。从机理上讲,RIOK3与热休克蛋白90α(HSP90α)相互作用,从而促进HSP90α与异柠檬酸脱氢酶1(IDH1)结合。这种相互作用会进一步上调 IDH1 的表达,促进 NADPH 的产生并保持氧化还原平衡。此外,抑制 RIOK3 对细胞内 NADPH 水平和 HSP90α 敲除细胞的细胞死亡率没有明显影响。总之,我们的研究结果表明,RIOK3 通过 HSP90α 依赖性途径维持结肠癌细胞在低糖环境中的存活。这凸显了RIOK3-HSP90α-IDH1级联的重要性,为在代谢应激条件下治疗结肠癌提供了潜在的靶向治疗策略。
{"title":"RIOK3 sustains colorectal cancer cell survival under glucose deprivation via an HSP90α-dependent pathway","authors":"Nan Zhang, Lu Dong, Tingting Ning, Feng Du, Mengran Zhao, Junxuan Xu, Sian Xie, Si Liu, Xiujing Sun, Peng Li, Shutian Zhang, Shengtao Zhu","doi":"10.1038/s41389-024-00514-5","DOIUrl":"https://doi.org/10.1038/s41389-024-00514-5","url":null,"abstract":"<p>Glucose oxidation via the pentose phosphate pathway serves as the primary cellular mechanism for generating nicotinamide adenine dinucleotide phosphate (NADPH). The central regions of solid tumors typically experience glucose deficiency, emphasizing the need for sustained NADPH production crucial to tumor cell survival. This study highlights the crucial role of RIOK3 in maintaining NADPH production and colorectal cancer (CRC) cell survival during glucose deficiency. Our findings revealed upregulated RIOK3 expression upon glucose deprivation, with RIOK3 knockout significantly reducing cancer cell survival. Mechanistically, RIOK3 interacts with heat shock protein 90α (HSP90α), a chaperone integral to various cellular processes, thereby facilitating HSP90α binding to isocitrate dehydrogenase 1 (IDH1). This interaction further upregulates IDH1 expression, enhancing NADPH production and preserving redox balance. Furthermore, RIOK3 inhibition had no discernible effect on intracellular NADPH levels and cell death rates in HSP90α-knockdown cells. Collectively, our findings suggest that RIOK3 sustains colon cancer cell survival in low-glucose environments through an HSP90α-dependent pathway. This highlights the significance of the RIOK3–HSP90α–IDH1 cascade, providing insights into potential targeted therapeutic strategies for CRC in metabolic stress conditions.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-03-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140057699","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Designing patient-oriented combination therapies for acute myeloid leukemia based on efficacy/toxicity integration and bipartite network modeling. 基于疗效/毒性整合和双向网络建模,设计以患者为导向的急性髓性白血病联合疗法。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-03-01 DOI: 10.1038/s41389-024-00510-9
Mehdi Mirzaie, Elham Gholizadeh, Juho J Miettinen, Filipp Ianevski, Tanja Ruokoranta, Jani Saarela, Mikko Manninen, Susanna Miettinen, Caroline A Heckman, Mohieddin Jafari

Acute myeloid leukemia (AML), a heterogeneous and aggressive blood cancer, does not respond well to single-drug therapy. A combination of drugs is required to effectively treat this disease. Computational models are critical for combination therapy discovery due to the tens of thousands of two-drug combinations, even with approved drugs. While predicting synergistic drugs is the focus of current methods, few consider drug efficacy and potential toxicity, which are crucial for treatment success. To find effective new drug candidates, we constructed a bipartite network using patient-derived tumor samples and drugs. The network is based on drug-response screening and summarizes all treatment response heterogeneity as drug response weights. This bipartite network is then projected onto the drug part, resulting in the drug similarity network. Distinct drug clusters were identified using community detection methods, each targeting different biological processes and pathways as revealed by enrichment and pathway analysis of the drugs' protein targets. Four drugs with the highest efficacy and lowest toxicity from each cluster were selected and tested for drug sensitivity using cell viability assays on various samples. Results show that ruxolitinib-ulixertinib and sapanisertib-LY3009120 are the most effective combinations with the least toxicity and the best synergistic effect on blast cells. These findings lay the foundation for personalized and successful AML therapies, ultimately leading to the development of drug combinations that can be used alongside standard first-line AML treatment.

急性髓性白血病(AML)是一种异质性侵袭性血癌,单药治疗效果不佳。要有效治疗这种疾病,需要联合用药。即使是已获批准的药物,也有数以万计的双药组合,因此计算模型对于发现联合疗法至关重要。虽然预测协同药物是当前方法的重点,但很少有方法会考虑药物疗效和潜在毒性,而这对治疗成功至关重要。为了找到有效的候选新药,我们利用源自患者的肿瘤样本和药物构建了一个双方网络。该网络基于药物反应筛选,并将所有治疗反应异质性总结为药物反应权重。然后将该双向网络投射到药物部分,形成药物相似性网络。通过对药物的蛋白质靶点进行富集和通路分析,发现了各自针对不同生物过程和通路的药物群集。从每个群组中选出了四种疗效最高、毒性最低的药物,并在不同样本上使用细胞活力测定法进行了药物敏感性测试。结果显示,ruxolitinib-ulixertinib和sapanisertib-LY3009120是最有效的组合,毒性最小,对爆破细胞的协同效应最好。这些发现为个性化和成功的急性髓细胞性白血病疗法奠定了基础,最终将开发出可与标准一线急性髓细胞性白血病治疗同时使用的药物组合。
{"title":"Designing patient-oriented combination therapies for acute myeloid leukemia based on efficacy/toxicity integration and bipartite network modeling.","authors":"Mehdi Mirzaie, Elham Gholizadeh, Juho J Miettinen, Filipp Ianevski, Tanja Ruokoranta, Jani Saarela, Mikko Manninen, Susanna Miettinen, Caroline A Heckman, Mohieddin Jafari","doi":"10.1038/s41389-024-00510-9","DOIUrl":"10.1038/s41389-024-00510-9","url":null,"abstract":"<p><p>Acute myeloid leukemia (AML), a heterogeneous and aggressive blood cancer, does not respond well to single-drug therapy. A combination of drugs is required to effectively treat this disease. Computational models are critical for combination therapy discovery due to the tens of thousands of two-drug combinations, even with approved drugs. While predicting synergistic drugs is the focus of current methods, few consider drug efficacy and potential toxicity, which are crucial for treatment success. To find effective new drug candidates, we constructed a bipartite network using patient-derived tumor samples and drugs. The network is based on drug-response screening and summarizes all treatment response heterogeneity as drug response weights. This bipartite network is then projected onto the drug part, resulting in the drug similarity network. Distinct drug clusters were identified using community detection methods, each targeting different biological processes and pathways as revealed by enrichment and pathway analysis of the drugs' protein targets. Four drugs with the highest efficacy and lowest toxicity from each cluster were selected and tested for drug sensitivity using cell viability assays on various samples. Results show that ruxolitinib-ulixertinib and sapanisertib-LY3009120 are the most effective combinations with the least toxicity and the best synergistic effect on blast cells. These findings lay the foundation for personalized and successful AML therapies, ultimately leading to the development of drug combinations that can be used alongside standard first-line AML treatment.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10907624/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140013100","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Progesterone receptor potentiates macropinocytosis through CDC42 in pancreatic ductal adenocarcinoma. 胰腺导管腺癌中的黄体酮受体通过 CDC42 强化大蛋白细胞增殖。
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-29 DOI: 10.1038/s41389-024-00512-7
Ying-Na Liao, Yan-Zhi Gai, Li-Heng Qian, Hong Pan, Yi-Fan Zhang, Pin Li, Ying Guo, Shu-Xin Li, Hui-Zhen Nie

Endocrine receptors play an essential role in tumor metabolic reprogramming and represent a promising therapeutic avenue in pancreatic ductal adenocarcinoma (PDAC). PDAC is characterized by a nutrient-deprived microenvironment. To meet their ascendant energy demands, cancer cells can internalize extracellular proteins via macropinocytosis. However, the roles of endocrine receptors in macropinocytosis are not clear. In this study, we found that progesterone receptor (PGR), a steroid-responsive nuclear receptor, is highly expressed in PDAC tissues obtained from both patients and transgenic LSL-KrasG12D/+; LSL-Trp53R172H/+; PDX1-cre (KPC) mice. Moreover, PGR knockdown restrained PDAC cell survival and tumor growth both in vitro and in vivo. Genetic and pharmacological PGR inhibition resulted in a marked attenuation of macropinocytosis in PDAC cells and subcutaneous tumor models, indicating the involvement of this receptor in macropinocytosis regulation. Mechanistically, PGR upregulated CDC42, a critical regulator in macropinocytosis, through PGR-mediated transcriptional activation. These data deepen the understanding of how the endocrine system influences tumor progression via a non-classical pathway and provide a novel therapeutic option for patients with PDAC.

内分泌受体在肿瘤代谢重编程中起着至关重要的作用,是治疗胰腺导管腺癌(PDAC)的有效途径。胰腺导管腺癌的特点是缺乏营养的微环境。为满足其不断上升的能量需求,癌细胞可通过大蛋白胞吞作用将细胞外蛋白质内化。然而,内分泌受体在大蛋白胞吞过程中的作用尚不明确。本研究发现,孕酮受体(PGR)是一种类固醇反应性核受体,在患者和转基因 LSL-KrasG12D/+; LSL-Trp53R172H/+; PDX1-cre (KPC) 小鼠的 PDAC 组织中均高表达。此外,PGR 基因敲除抑制了 PDAC 细胞在体外和体内的存活和肿瘤生长。基因和药物抑制 PGR 会导致 PDAC 细胞和皮下肿瘤模型中的大鼠胞吞功能明显减弱,这表明该受体参与了大鼠胞吞功能的调控。从机理上讲,PGR 通过 PGR 介导的转录激活,上调了大磷细胞吞噬过程中的关键调控因子 CDC42。这些数据加深了人们对内分泌系统如何通过非经典途径影响肿瘤进展的理解,并为 PDAC 患者提供了一种新的治疗选择。
{"title":"Progesterone receptor potentiates macropinocytosis through CDC42 in pancreatic ductal adenocarcinoma.","authors":"Ying-Na Liao, Yan-Zhi Gai, Li-Heng Qian, Hong Pan, Yi-Fan Zhang, Pin Li, Ying Guo, Shu-Xin Li, Hui-Zhen Nie","doi":"10.1038/s41389-024-00512-7","DOIUrl":"10.1038/s41389-024-00512-7","url":null,"abstract":"<p><p>Endocrine receptors play an essential role in tumor metabolic reprogramming and represent a promising therapeutic avenue in pancreatic ductal adenocarcinoma (PDAC). PDAC is characterized by a nutrient-deprived microenvironment. To meet their ascendant energy demands, cancer cells can internalize extracellular proteins via macropinocytosis. However, the roles of endocrine receptors in macropinocytosis are not clear. In this study, we found that progesterone receptor (PGR), a steroid-responsive nuclear receptor, is highly expressed in PDAC tissues obtained from both patients and transgenic LSL-Kras<sup>G12D/+</sup>; LSL-Trp53<sup>R172H/+</sup>; PDX1-cre (KPC) mice. Moreover, PGR knockdown restrained PDAC cell survival and tumor growth both in vitro and in vivo. Genetic and pharmacological PGR inhibition resulted in a marked attenuation of macropinocytosis in PDAC cells and subcutaneous tumor models, indicating the involvement of this receptor in macropinocytosis regulation. Mechanistically, PGR upregulated CDC42, a critical regulator in macropinocytosis, through PGR-mediated transcriptional activation. These data deepen the understanding of how the endocrine system influences tumor progression via a non-classical pathway and provide a novel therapeutic option for patients with PDAC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-02-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10904380/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139997026","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PKC-independent PI3K signalling diminishes PKC inhibitor sensitivity in uveal melanoma 不依赖 PKC 的 PI3K 信号降低了葡萄膜黑色素瘤对 PKC 抑制剂的敏感性
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-28 DOI: 10.1038/s41389-024-00511-8
John J. Park, Sabine Abou Hamad, Ashleigh Stewart, Matteo S. Carlino, Su Yin Lim, Helen Rizos

Protein kinase C (PKC) is activated downstream of gain-of-function GNAQ or GNA11 (GNAQ/GNA11) mutations in over 90% of uveal melanoma (UM). Phase I clinical trials of PKC inhibitors have shown modest response rates with no survival benefit in metastatic UM. Although PKC inhibitors actively suppress mitogen-activated protein kinase (MAPK) signalling in UM, the effect on other UM signalling cascades is not well understood. We examined the transcriptome of UM biopsies collected pre- and post-PKC inhibitor therapy and confirmed that MAPK, but not PI3K/AKT signalling, was inhibited early during treatment with the second-generation PKC inhibitor IDE196. Similarly, in GNAQ/GNA11-mutant UM cell models, PKC inhibitor monotherapy effectively suppressed MAPK activity, but PI3K/AKT signalling remained active, and thus, concurrent inhibition of PKC and PI3K/AKT signalling was required to synergistically induce cell death in a panel of GNAQ/GNA11-mutant UM cell lines. We also show that re-activation of MAPK signalling has a dominant role in regulating PKC inhibitor responses in UM and that PI3K/AKT signalling diminishes UM cell sensitivity to PKC inhibitor monotherapy. Thus, combination therapies targeting PKC and PKC-independent signalling nodes, including PI3K/AKT activity, are required to improve responses in patients with metastatic UM.

在超过90%的葡萄膜黑色素瘤(UM)中,蛋白激酶C(PKC)在功能增益型GNAQ或GNA11(GNAQ/GNA11)突变的下游被激活。PKC 抑制剂的 I 期临床试验显示,转移性 UM 的反应率不高,但无生存获益。虽然PKC抑制剂能积极抑制UM中的丝裂原活化蛋白激酶(MAPK)信号传导,但对UM其他信号级联的影响尚不十分清楚。我们研究了在 PKC 抑制剂治疗前后收集的 UM 活检组织的转录组,结果证实,在使用第二代 PKC 抑制剂 IDE196 治疗的早期,MAPK(而非 PI3K/AKT 信号)受到抑制。同样,在 GNAQ/GNA11 突变 UM 细胞模型中,PKC 抑制剂单药治疗能有效抑制 MAPK 活性,但 PI3K/AKT 信号仍处于活跃状态,因此,在一组 GNAQ/GNA11 突变 UM 细胞系中,需要同时抑制 PKC 和 PI3K/AKT 信号才能协同诱导细胞死亡。我们还发现,MAPK 信号的重新激活在调节 UM 对 PKC 抑制剂的反应中起着主导作用,而 PI3K/AKT 信号会降低 UM 细胞对 PKC 抑制剂单一疗法的敏感性。因此,需要针对 PKC 和 PKC 依赖性信号节点(包括 PI3K/AKT 活性)的联合疗法来改善转移性 UM 患者的反应。
{"title":"PKC-independent PI3K signalling diminishes PKC inhibitor sensitivity in uveal melanoma","authors":"John J. Park, Sabine Abou Hamad, Ashleigh Stewart, Matteo S. Carlino, Su Yin Lim, Helen Rizos","doi":"10.1038/s41389-024-00511-8","DOIUrl":"https://doi.org/10.1038/s41389-024-00511-8","url":null,"abstract":"<p>Protein kinase C (PKC) is activated downstream of gain-of-function <i>GNAQ</i> or <i>GNA11</i> (<i>GNAQ/GNA11</i>) mutations in over 90% of uveal melanoma (UM). Phase I clinical trials of PKC inhibitors have shown modest response rates with no survival benefit in metastatic UM. Although PKC inhibitors actively suppress mitogen-activated protein kinase (MAPK) signalling in UM, the effect on other UM signalling cascades is not well understood. We examined the transcriptome of UM biopsies collected pre- and post-PKC inhibitor therapy and confirmed that MAPK, but not PI3K/AKT signalling, was inhibited early during treatment with the second-generation PKC inhibitor IDE196. Similarly, in GNAQ/GNA11-mutant UM cell models, PKC inhibitor monotherapy effectively suppressed MAPK activity, but PI3K/AKT signalling remained active, and thus, concurrent inhibition of PKC and PI3K/AKT signalling was required to synergistically induce cell death in a panel of GNAQ/GNA11-mutant UM cell lines. We also show that re-activation of MAPK signalling has a dominant role in regulating PKC inhibitor responses in UM and that PI3K/AKT signalling diminishes UM cell sensitivity to PKC inhibitor monotherapy. Thus, combination therapies targeting PKC and PKC-independent signalling nodes, including PI3K/AKT activity, are required to improve responses in patients with metastatic UM.</p><figure></figure>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-02-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139987822","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DDX3X interacts with SIRT7 to promote PD-L1 expression to facilitate PDAC progression DDX3X 与 SIRT7 相互作用,促进 PD-L1 的表达,从而推动 PDAC 的进展
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-02-05 DOI: 10.1038/s41389-024-00509-2
Tianming Zhao, Hanlong Zhu, Tianhui Zou, Si Zhao, Lin Zhou, Muhan Ni, Feng Liu, Hao Zhu, Xiaotan Dou, Jian Di, Bing Xu, Lei Wang, Xiaoping Zou

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is recognized as the most aggressive and fatal malignancy. A previous study reported that PDAC patients who exhibit elevated levels of DDX3X have a poor prognosis and low overall survival rate. However, the underlying molecular mechanism remains unclear. This study aimed to investigate the specific roles of DDX3X in PDAC. Multiple bioinformatics analyses were used to evaluate DDX3X expression and its potential role in PDAC. In vitro and in vivo studies were performed to assess the effects of DDX3X on PDAC cell growth. Furthermore, Western blotting, quantitative PCR, immunohistochemistry, immunofluorescence, mass spectrometry, coimmunoprecipitation and multiplexed immunohistochemical staining were conducted to identify the specific regulatory mechanism in PDAC. The results verified that DDX3X expression is notably upregulated in the tumor tissue vs. normal tissue of PDAC patients. DDX3X knockdown markedly suppressed the proliferation, invasion and migration of PDAC cells in vitro and inhibited tumor growth in vivo. Conversely, overexpression of DDX3X induced the opposite effect. Further studies supported that the DDX3X protein can associate with sirtuin 7 (SIRT7) to stimulate PDAC carcinogenesis and progression. Furthermore, SIRT7 inhibition significantly impeded DDX3X-mediated tumor growth both ex vivo and in vivo. The results also revealed that programmed death ligand 1 (PD-L1) expression is positively correlated with DDX3X expression. These results reveal significant involvement of the DDX3X-SIRT7 axis in the initiation and advancement of PDAC and offer previously undiscovered therapeutic options for PDAC management.

摘要 胰腺导管腺癌(PDAC)被认为是最具侵袭性和致命性的恶性肿瘤。先前的一项研究报告显示,DDX3X水平升高的PDAC患者预后较差,总生存率较低。然而,其潜在的分子机制仍不清楚。本研究旨在探讨 DDX3X 在 PDAC 中的特定作用。研究人员使用多种生物信息学分析评估了DDX3X的表达及其在PDAC中的潜在作用。体外和体内研究评估了 DDX3X 对 PDAC 细胞生长的影响。此外,还进行了 Western 印迹、定量 PCR、免疫组化、免疫荧光、质谱分析、共沉淀和多重免疫组化染色,以确定 PDAC 中的特定调控机制。结果证实,与正常组织相比,DDX3X在PDAC患者的肿瘤组织中表达明显上调。敲除 DDX3X 能显著抑制 PDAC 细胞在体外的增殖、侵袭和迁移,并抑制肿瘤在体内的生长。相反,过表达 DDX3X 则会引起相反的效果。进一步的研究证实,DDX3X 蛋白可与 sirtuin 7(SIRT7)结合,刺激 PDAC 癌变和进展。此外,抑制 SIRT7 能显著抑制 DDX3X 介导的体内外肿瘤生长。研究结果还发现,程序性死亡配体1(PD-L1)的表达与DDX3X的表达呈正相关。这些结果揭示了 DDX3X-SIRT7 轴在 PDAC 的发生和发展过程中的重要作用,并为 PDAC 的治疗提供了之前尚未发现的治疗方案。
{"title":"DDX3X interacts with SIRT7 to promote PD-L1 expression to facilitate PDAC progression","authors":"Tianming Zhao, Hanlong Zhu, Tianhui Zou, Si Zhao, Lin Zhou, Muhan Ni, Feng Liu, Hao Zhu, Xiaotan Dou, Jian Di, Bing Xu, Lei Wang, Xiaoping Zou","doi":"10.1038/s41389-024-00509-2","DOIUrl":"https://doi.org/10.1038/s41389-024-00509-2","url":null,"abstract":"<h3 data-test=\"abstract-sub-heading\">Abstract</h3><p>Pancreatic ductal adenocarcinoma (PDAC) is recognized as the most aggressive and fatal malignancy. A previous study reported that PDAC patients who exhibit elevated levels of DDX3X have a poor prognosis and low overall survival rate. However, the underlying molecular mechanism remains unclear. This study aimed to investigate the specific roles of DDX3X in PDAC. Multiple bioinformatics analyses were used to evaluate DDX3X expression and its potential role in PDAC. In vitro and in vivo studies were performed to assess the effects of DDX3X on PDAC cell growth. Furthermore, Western blotting, quantitative PCR, immunohistochemistry, immunofluorescence, mass spectrometry, coimmunoprecipitation and multiplexed immunohistochemical staining were conducted to identify the specific regulatory mechanism in PDAC. The results verified that DDX3X expression is notably upregulated in the tumor tissue vs. normal tissue of PDAC patients. DDX3X knockdown markedly suppressed the proliferation, invasion and migration of PDAC cells in vitro and inhibited tumor growth in vivo. Conversely, overexpression of DDX3X induced the opposite effect. Further studies supported that the DDX3X protein can associate with sirtuin 7 (SIRT7) to stimulate PDAC carcinogenesis and progression. Furthermore, SIRT7 inhibition significantly impeded DDX3X-mediated tumor growth both ex vivo and in vivo. The results also revealed that programmed death ligand 1 (PD-L1) expression is positively correlated with DDX3X expression. These results reveal significant involvement of the DDX3X-SIRT7 axis in the initiation and advancement of PDAC and offer previously undiscovered therapeutic options for PDAC management.</p><figure></figure>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139688969","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
LUBAC promotes angiogenesis and lung tumorigenesis by ubiquitinating and antagonizing autophagic degradation of HIF1α LUBAC 通过泛素化和拮抗 HIF1α 的自噬降解促进血管生成和肺肿瘤发生
IF 6.2 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-25 DOI: 10.1038/s41389-024-00508-3
Ying Jin, Yazhi Peng, Jie Xu, Ye Yuan, Nan Yang, Zemei Zhang, Lei Xu, Lin Li, Yulian Xiong, Dejiao Sun, Yamu Pan, Ruiqing Wu, Jian Fu

Hypoxia-inducible factor 1 (HIF1) is critically important for driving angiogenesis and tumorigenesis. Linear ubiquitin chain assembly complex (LUBAC), the only known ubiquitin ligase capable of catalyzing protein linear ubiquitination to date, is implicated in cell signaling and associated with cancers. However, the role and mechanism of LUBAC in regulating the expression and function of HIF1α, the labile subunit of HIF1, remain to be elucidated. Herein we showed that LUBAC increases HIF1α protein expression in cultured cells and tissues of human lung cancer and enhances HIF1α DNA-binding and transcriptional activities, which are dependent upon LUBAC enzymatic activity. Mechanistically, LUBAC increases HIF1α stability through antagonizing HIF1α decay by the chaperone-mediated autophagy (CMA)-lysosome pathway, thereby potentiating HIF1α activity. We further demonstrated that HIF1α selectively interacts with HOIP (the catalytic subunit of LUBAC) primarily in the cytoplasm. LUBAC catalyzes linear ubiquitination of HIF1α at lysine 362. Linear ubiquitination shields HIF1α from interacting with heat-shock cognate protein of 70 kDa and lysosome-associated membrane protein type 2 A, two components of CMA. Consequently, linear ubiquitination confers protection against CMA-mediated destruction of HIF1α, increasing HIF1α stability and activity. We found that prolyl hydroxylation is not a perquisite for LUBAC’s effects on HIF1α. Functionally, LUBAC facilitates proliferation, clonogenic formation, invasion and migration of lung cancer cells. LUBAC also boosts angiogenesis and exacerbates lung cancer growth in mice, which are greatly compromised by inhibition of HIF1α. This work provides novel mechanistic insights into the role of LUBAC in regulating HIF1α homeostasis, tumor angiogenesis and tumorigenesis of lung cancer, making LUBAC an attractive therapeutic target for cancers.

缺氧诱导因子 1(HIF1)对血管生成和肿瘤发生具有至关重要的推动作用。线性泛素链组装复合物(LUBAC)是迄今所知唯一能够催化蛋白质线性泛素化的泛素连接酶,它与细胞信号传导有关,并与癌症相关。然而,LUBAC 在调节 HIF1 的易变亚基 HIF1α 的表达和功能方面的作用和机制仍有待阐明。在本文中,我们发现 LUBAC 可增加培养细胞和人类肺癌组织中 HIF1α 蛋白的表达,并增强 HIF1α DNA 结合和转录活性,而这些都依赖于 LUBAC 的酶活性。从机理上讲,LUBAC通过拮抗伴侣介导的自噬(CMA)-溶酶体途径的HIF1α衰变,增加了HIF1α的稳定性,从而增强了HIF1α的活性。我们进一步证实,HIF1α 主要在细胞质中选择性地与 HOIP(LUBAC 的催化亚基)相互作用。LUBAC 催化 HIF1α 赖氨酸 362 处的线性泛素化。线性泛素化使 HIF1α 无法与 70 kDa 的热休克认知蛋白和溶酶体相关膜蛋白 2 A 型(CMA 的两个组成部分)相互作用。因此,线性泛素化能防止 CMA 介导的对 HIF1α 的破坏,提高 HIF1α 的稳定性和活性。我们发现,脯氨酰羟基化并不是 LUBAC 对 HIF1α 起作用的先决条件。在功能上,LUBAC 可促进肺癌细胞的增殖、克隆形成、侵袭和迁移。LUBAC 还能促进血管生成并加剧小鼠的肺癌生长,而抑制 HIF1α 则会大大降低这些作用。这项研究从机理上揭示了LUBAC在调节HIF1α平衡、肿瘤血管生成和肺癌肿瘤发生中的作用,使LUBAC成为一个有吸引力的癌症治疗靶点。
{"title":"LUBAC promotes angiogenesis and lung tumorigenesis by ubiquitinating and antagonizing autophagic degradation of HIF1α","authors":"Ying Jin, Yazhi Peng, Jie Xu, Ye Yuan, Nan Yang, Zemei Zhang, Lei Xu, Lin Li, Yulian Xiong, Dejiao Sun, Yamu Pan, Ruiqing Wu, Jian Fu","doi":"10.1038/s41389-024-00508-3","DOIUrl":"https://doi.org/10.1038/s41389-024-00508-3","url":null,"abstract":"<p>Hypoxia-inducible factor 1 (HIF1) is critically important for driving angiogenesis and tumorigenesis. Linear ubiquitin chain assembly complex (LUBAC), the only known ubiquitin ligase capable of catalyzing protein linear ubiquitination to date, is implicated in cell signaling and associated with cancers. However, the role and mechanism of LUBAC in regulating the expression and function of HIF1α, the labile subunit of HIF1, remain to be elucidated. Herein we showed that LUBAC increases HIF1α protein expression in cultured cells and tissues of human lung cancer and enhances HIF1α DNA-binding and transcriptional activities, which are dependent upon LUBAC enzymatic activity. Mechanistically, LUBAC increases HIF1α stability through antagonizing HIF1α decay by the chaperone-mediated autophagy (CMA)-lysosome pathway, thereby potentiating HIF1α activity. We further demonstrated that HIF1α selectively interacts with HOIP (the catalytic subunit of LUBAC) primarily in the cytoplasm. LUBAC catalyzes linear ubiquitination of HIF1α at lysine 362. Linear ubiquitination shields HIF1α from interacting with heat-shock cognate protein of 70 kDa and lysosome-associated membrane protein type 2 A, two components of CMA. Consequently, linear ubiquitination confers protection against CMA-mediated destruction of HIF1α, increasing HIF1α stability and activity. We found that prolyl hydroxylation is not a perquisite for LUBAC’s effects on HIF1α. Functionally, LUBAC facilitates proliferation, clonogenic formation, invasion and migration of lung cancer cells. LUBAC also boosts angiogenesis and exacerbates lung cancer growth in mice, which are greatly compromised by inhibition of HIF1α. This work provides novel mechanistic insights into the role of LUBAC in regulating HIF1α homeostasis, tumor angiogenesis and tumorigenesis of lung cancer, making LUBAC an attractive therapeutic target for cancers.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":6.2,"publicationDate":"2024-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139552430","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncogenesis
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