首页 > 最新文献

Cellular Oncology最新文献

英文 中文
Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells. 肿瘤微环境的酸度和缺氧是肿瘤细胞释放细胞外囊泡的积极相互作用。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2025-02-01 Epub Date: 2024-07-18 DOI: 10.1007/s13402-024-00969-z
Silvia Peppicelli, Lido Calorini, Francesca Bianchini, Laura Papucci, Lucia Magnelli, Elena Andreucci

The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.

肿瘤微环境的特征复杂且不断变化,因肿瘤组织类型而异,其特点是宿主细胞和肿瘤细胞嵌入缺氧和低 pH 值的环境中,这是血管和肿瘤细胞增殖之间经常失衡的结果。这些微环境代谢压力因素在宿主细胞和肿瘤细胞的重塑过程中发挥着至关重要的作用,有助于刺激癌细胞的异质性、克隆进化和多药耐药性,最终导致癌细胞的发展和转移。细胞外囊泡(EVs)是肿瘤/宿主细胞释放到细胞外环境中的膜封闭结构,现已被认为是缺氧/酸性微环境中肿瘤细胞与当地细胞成分之间复杂的细胞间交流的关键驱动因素。了解由 EVs 释放引发的缺氧和酸性微环境中肿瘤细胞与宿主细胞之间相互作用的复杂分子机制,可为制定创新策略以破坏癌细胞与其微环境之间的复杂相互作用铺平道路。这种方法可能有助于开发一种高效、安全的治疗策略来对抗癌症进展。因此,我们回顾了在缺氧/酸性肿瘤微环境中释放 EVs 的主要发现,以了解 EVs 在肿瘤向转移性疾病发展过程中的作用。
{"title":"Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells.","authors":"Silvia Peppicelli, Lido Calorini, Francesca Bianchini, Laura Papucci, Lucia Magnelli, Elena Andreucci","doi":"10.1007/s13402-024-00969-z","DOIUrl":"10.1007/s13402-024-00969-z","url":null,"abstract":"<p><p>The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"27-41"},"PeriodicalIF":6.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850579/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141632766","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
Antitumor activity of afatinib in EGFR T790M-negative human oral cancer therapeutically targets mTOR/Mcl-1 signaling axis. 阿法替尼对表皮生长因子受体(EGFR)T790M阴性人类口腔癌的抗肿瘤活性以mTOR/Mcl-1信号轴为治疗靶点。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2025-02-01 Epub Date: 2024-06-18 DOI: 10.1007/s13402-024-00962-6
Jung-Min Han, Kyu-Young Oh, Su-Jung Choi, Won-Woo Lee, Bo-Hwan Jin, Ji-Hoon Kim, Hyun-Ju Yu, Ryan Jin Young Kim, Hye-Jung Yoon, Jae-Il Lee, Seong-Doo Hong, Sung-Dae Cho

Purpose: This study investigates the role and effectiveness of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) in oral cancer, focusing on the clinical relevance of EGFR and myeloid cell leukemia-1 (Mcl-1) in head and neck cancers (HNCs). It aims to explore the molecular mechanism of afatinib, a TKI, in treating human oral cancer.

Methods: We conducted an in silico analysis using databases like The Cancer Genome Atlas, Gene Expression Omnibus, and Clinical Proteomic Tumor Analysis Consortium, along with immunohistochemistry staining, to study EGFR and Mcl-1 expression in HNCs. For investigating afatinib's anticancer properties, we performed various in vitro and in vivo analyses, including trypan blue exclusion assay, Western blotting, 4'-6-diamidino-2-phenylindole staining, flow cytometry, quantitative real-time PCR, Mitochondrial membrane potential assay, overexpression vector construction, transient transfection, and a tumor xenograft model.

Results: Higher expression levels of EGFR and Mcl-1 were observed in HNC patient tissues compared to normal tissues, with their co-expression significantly linked to poor prognosis. There was a strong correlation between EGFR and Mcl-1 expressions in oral cancer patients. Afatinib treatment induced apoptosis and suppressed Mcl-1 in oral cancer cell lines without the EGFR T790M mutation. The mechanism of afatinib-induced apoptosis involved the EGFR/mTOR/Mcl-1 axis, as shown by the effects of mTOR activator MHY1485 and inhibitor rapamycin. Afatinib also increased Bim expression, mitochondrial membrane permeabilization, and cytochrome c release. It significantly lowered tumor volume without affecting body, liver, and kidney weights.

Conclusion: Afatinib, targeting the EGFR/mTOR/Mcl-1 axis, shows promise as a therapeutic strategy for oral cancer, especially in patients with high EGFR and Mcl-1 expressions.

目的:本研究探讨表皮生长因子受体(EGFR)酪氨酸激酶抑制剂(TKI)在口腔癌中的作用和有效性,重点关注EGFR和髓细胞白血病-1(Mcl-1)在头颈癌(HNC)中的临床相关性。本研究旨在探索阿法替尼(一种TKI)治疗人类口腔癌的分子机制:我们利用癌症基因组图谱(The Cancer Genome Atlas)、基因表达总库(Gene Expression Omnibus)和临床蛋白质组肿瘤分析联盟(Clinical Proteomic Tumor Analysis Consortium)等数据库,结合免疫组化染色,对HNCs中表皮生长因子受体(EGFR)和Mcl-1的表达进行了硅学分析。为了研究阿法替尼的抗癌特性,我们进行了各种体内外分析,包括胰蓝排除试验、Western印迹、4'-6-二脒基-2-苯基吲哚染色、流式细胞术、定量实时PCR、线粒体膜电位检测、过表达载体构建、瞬时转染和肿瘤异种移植模型:结果:与正常组织相比,HNC患者组织中表皮生长因子受体(EGFR)和Mcl-1的表达水平更高,它们的共同表达与预后不良密切相关。在口腔癌患者中,表皮生长因子受体(EGFR)和Mcl-1的表达有很强的相关性。阿法替尼治疗可诱导未发生表皮生长因子受体T790M突变的口腔癌细胞株凋亡并抑制Mcl-1。阿法替尼诱导细胞凋亡的机制涉及表皮生长因子受体/mTOR/Mcl-1轴,mTOR激活剂MHY1485和抑制剂雷帕霉素的作用也证明了这一点。阿法替尼还能增加Bim的表达、线粒体膜的通透性和细胞色素c的释放。阿法替尼能明显降低肿瘤体积,而不影响体重、肝脏和肾脏重量:结论:以表皮生长因子受体/mTOR/Mcl-1轴为靶点的阿法替尼有望成为口腔癌的一种治疗策略,尤其是在表皮生长因子受体和Mcl-1高表达的患者中。
{"title":"Antitumor activity of afatinib in EGFR T790M-negative human oral cancer therapeutically targets mTOR/Mcl-1 signaling axis.","authors":"Jung-Min Han, Kyu-Young Oh, Su-Jung Choi, Won-Woo Lee, Bo-Hwan Jin, Ji-Hoon Kim, Hyun-Ju Yu, Ryan Jin Young Kim, Hye-Jung Yoon, Jae-Il Lee, Seong-Doo Hong, Sung-Dae Cho","doi":"10.1007/s13402-024-00962-6","DOIUrl":"10.1007/s13402-024-00962-6","url":null,"abstract":"<p><strong>Purpose: </strong>This study investigates the role and effectiveness of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) in oral cancer, focusing on the clinical relevance of EGFR and myeloid cell leukemia-1 (Mcl-1) in head and neck cancers (HNCs). It aims to explore the molecular mechanism of afatinib, a TKI, in treating human oral cancer.</p><p><strong>Methods: </strong>We conducted an in silico analysis using databases like The Cancer Genome Atlas, Gene Expression Omnibus, and Clinical Proteomic Tumor Analysis Consortium, along with immunohistochemistry staining, to study EGFR and Mcl-1 expression in HNCs. For investigating afatinib's anticancer properties, we performed various in vitro and in vivo analyses, including trypan blue exclusion assay, Western blotting, 4'-6-diamidino-2-phenylindole staining, flow cytometry, quantitative real-time PCR, Mitochondrial membrane potential assay, overexpression vector construction, transient transfection, and a tumor xenograft model.</p><p><strong>Results: </strong>Higher expression levels of EGFR and Mcl-1 were observed in HNC patient tissues compared to normal tissues, with their co-expression significantly linked to poor prognosis. There was a strong correlation between EGFR and Mcl-1 expressions in oral cancer patients. Afatinib treatment induced apoptosis and suppressed Mcl-1 in oral cancer cell lines without the EGFR T790M mutation. The mechanism of afatinib-induced apoptosis involved the EGFR/mTOR/Mcl-1 axis, as shown by the effects of mTOR activator MHY1485 and inhibitor rapamycin. Afatinib also increased Bim expression, mitochondrial membrane permeabilization, and cytochrome c release. It significantly lowered tumor volume without affecting body, liver, and kidney weights.</p><p><strong>Conclusion: </strong>Afatinib, targeting the EGFR/mTOR/Mcl-1 axis, shows promise as a therapeutic strategy for oral cancer, especially in patients with high EGFR and Mcl-1 expressions.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"123-138"},"PeriodicalIF":6.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850456/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141418099","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
Suppressing SENP1 inhibits esophageal squamous carcinoma cell growth via SIRT6 SUMOylation. 抑制 SENP1 可通过 SIRT6 SUMOylation 抑制食管鳞癌细胞生长
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2025-02-01 Epub Date: 2024-07-02 DOI: 10.1007/s13402-024-00956-4
Jianmin Gu, Shaoyuan Zhang, Dong Lin, Wenhan Wang, Jinke Cheng, Quan Zheng, Hao Wang, Lijie Tan

Purpose: Esophageal squamous cell carcinoma (ESCC) is a prevalent tumor in the gastrointestinal tract, but our understanding of the molecular mechanisms underlying ESCC remains incomplete. Existing studies indicate that SUMO specific peptidase 1 (SENP1) plays a crucial role in the development and progression of various malignant tumors through diverse molecular mechanisms. However, the functional mechanism and clinical implications of SENP1 in the progression of ESCC remain unclear.

Methods: Bulk RNA-Sequencing (RNA-seq) was used to compare potential genes in the esophageal tissues of mice with ESCC to the control group. The up-regulated SENP1 was selected. The protein level of SENP1 in ESCC patient samples was analyzed by immunohistochemistry and western blot. The potential prognostic value of SENP1 on overall survival of ESCC patients was examined using tissue microarray analysis and the Kaplan-Meier method. The biological function was confirmed through in vitro and in vivo knockdown approaches of SENP1. The role of SENP1 in cell cycle progression and apoptosis of ESCC cells was analyzed by flow cytometry and western blot. The downstream signaling pathways regulated by SENP1 were investigated via using RNA-Seq. SENP1-associated proteins were identified through immunoprecipitation. Overexpression of Sirtuin 6 (SIRT6) wildtype and mutant was performed to investigate the regulatory role of SENP1 in ESCC progression in vitro.

Results: Our study discovered that SENP1 was upregulated in ESCC tissues and served as a novel prognostic factor. Moreover, SENP1 enhanced cell proliferation and migration of ESCC cell lines in vitro, as well as promoted tumor growth in vivo. Thymidine kinase 1 (TK1), Geminin (GMNN), cyclin dependent kinase 1(CDK1), and cyclin A2 (CCNA2) were identified as downstream genes of SENP1. Mechanistically, SENP1 deSUMOylated SIRT6 and subsequently inhibited SIRT6-mediated histone 3 lysine 56 (H3K56) deacetylation on those downstream genes. SIRT6 SUMOylation mutant (4KR) rescued the growth inhibition upon SENP1 depletion.

Conclusions: SENP1 promotes the malignant progression of ESCC by inhibiting the deacetylase activity of SIRT6 pathway through deSUMOylation. Our findings suggest that SENP1 may serve as a valuable biomarker for prognosis and a target for therapeutic intervention in ESCC.

目的:食管鳞状细胞癌(ESCC)是胃肠道中的一种常见肿瘤,但我们对其分子机制的了解仍不全面。现有研究表明,SUMO 特异性肽酶 1(SENP1)通过不同的分子机制在各种恶性肿瘤的发生和发展过程中起着至关重要的作用。然而,SENP1在ESCC进展过程中的功能机制和临床意义仍不清楚:方法:采用大量 RNA 序列分析(RNA-seq)方法比较了 ESCC 小鼠食管组织与对照组中的潜在基因。筛选出上调的 SENP1。通过免疫组化和免疫印迹分析了 ESCC 患者样本中 SENP1 的蛋白水平。利用组织芯片分析和 Kaplan-Meier 法研究了 SENP1 对 ESCC 患者总生存期的潜在预后价值。通过体外和体内敲除 SENP1 的方法证实了其生物学功能。流式细胞术和 Western 印迹分析了 SENP1 在 ESCC 细胞周期进展和凋亡中的作用。通过RNA-Seq研究了SENP1调控的下游信号通路。通过免疫沉淀鉴定了 SENP1 相关蛋白。过表达Sirtuin 6(SIRT6)野生型和突变型,研究SENP1在ESCC体外进展中的调控作用:结果:我们的研究发现,SENP1在ESCC组织中上调,是一种新的预后因子。此外,SENP1 在体外可增强 ESCC 细胞系的细胞增殖和迁移,在体内可促进肿瘤生长。研究发现胸苷激酶1(TK1)、Geminin(GMNN)、细胞周期蛋白依赖性激酶1(CDK1)和细胞周期蛋白A2(CCNA2)是SENP1的下游基因。从机理上讲,SENP1 对 SIRT6 进行去 SUMOO 化,从而抑制了 SIRT6 介导的组蛋白 3 赖氨酸 56(H3K56)在这些下游基因上的去乙酰化。SIRT6 SUMOylation突变体(4KR)能挽救SENP1耗竭后的生长抑制作用:结论:SENP1通过去SUMOylation抑制SIRT6通路的去乙酰化酶活性,从而促进ESCC的恶性进展。我们的研究结果表明,SENP1 可作为 ESCC 有价值的预后生物标志物和治疗干预靶点。
{"title":"Suppressing SENP1 inhibits esophageal squamous carcinoma cell growth via SIRT6 SUMOylation.","authors":"Jianmin Gu, Shaoyuan Zhang, Dong Lin, Wenhan Wang, Jinke Cheng, Quan Zheng, Hao Wang, Lijie Tan","doi":"10.1007/s13402-024-00956-4","DOIUrl":"10.1007/s13402-024-00956-4","url":null,"abstract":"<p><strong>Purpose: </strong>Esophageal squamous cell carcinoma (ESCC) is a prevalent tumor in the gastrointestinal tract, but our understanding of the molecular mechanisms underlying ESCC remains incomplete. Existing studies indicate that SUMO specific peptidase 1 (SENP1) plays a crucial role in the development and progression of various malignant tumors through diverse molecular mechanisms. However, the functional mechanism and clinical implications of SENP1 in the progression of ESCC remain unclear.</p><p><strong>Methods: </strong>Bulk RNA-Sequencing (RNA-seq) was used to compare potential genes in the esophageal tissues of mice with ESCC to the control group. The up-regulated SENP1 was selected. The protein level of SENP1 in ESCC patient samples was analyzed by immunohistochemistry and western blot. The potential prognostic value of SENP1 on overall survival of ESCC patients was examined using tissue microarray analysis and the Kaplan-Meier method. The biological function was confirmed through in vitro and in vivo knockdown approaches of SENP1. The role of SENP1 in cell cycle progression and apoptosis of ESCC cells was analyzed by flow cytometry and western blot. The downstream signaling pathways regulated by SENP1 were investigated via using RNA-Seq. SENP1-associated proteins were identified through immunoprecipitation. Overexpression of Sirtuin 6 (SIRT6) wildtype and mutant was performed to investigate the regulatory role of SENP1 in ESCC progression in vitro.</p><p><strong>Results: </strong>Our study discovered that SENP1 was upregulated in ESCC tissues and served as a novel prognostic factor. Moreover, SENP1 enhanced cell proliferation and migration of ESCC cell lines in vitro, as well as promoted tumor growth in vivo. Thymidine kinase 1 (TK1), Geminin (GMNN), cyclin dependent kinase 1(CDK1), and cyclin A2 (CCNA2) were identified as downstream genes of SENP1. Mechanistically, SENP1 deSUMOylated SIRT6 and subsequently inhibited SIRT6-mediated histone 3 lysine 56 (H3K56) deacetylation on those downstream genes. SIRT6 SUMOylation mutant (4KR) rescued the growth inhibition upon SENP1 depletion.</p><p><strong>Conclusions: </strong>SENP1 promotes the malignant progression of ESCC by inhibiting the deacetylase activity of SIRT6 pathway through deSUMOylation. Our findings suggest that SENP1 may serve as a valuable biomarker for prognosis and a target for therapeutic intervention in ESCC.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"67-81"},"PeriodicalIF":6.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850494/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141491025","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
Fasting in combination with the cocktail Sorafenib:Metformin blunts cellular plasticity and promotes liver cancer cell death via poly-metabolic exhaustion. 禁食与索拉非尼:二甲双胍鸡尾酒结合使用,会削弱细胞的可塑性,并通过多代谢衰竭促进肝癌细胞死亡。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2025-02-01 Epub Date: 2024-07-11 DOI: 10.1007/s13402-024-00966-2
Juan L López-Cánovas, Beatriz Naranjo-Martínez, Alberto Diaz-Ruiz

Purpose: Dual-Interventions targeting glucose and oxidative metabolism are receiving increasing attention in cancer therapy. Sorafenib (S) and Metformin (M), two gold-standards in liver cancer, are known for their mitochondrial inhibitory capacity. Fasting, a glucose-limiting strategy, is also emerging as chemotherapy adjuvant. Herein, we explore the anti-carcinogenic response of nutrient restriction in combination with sorafenib:metformin (NR-S:M).

Results: Our data demonstrates that, independently of liver cancer aggressiveness, fasting synergistically boosts the anti-proliferative effects of S:M co-treatment. Metabolic and Cellular plasticity was determined by the examination of mitochondrial and glycolytic activity, cell cycle modulation, activation of cellular apoptosis, and regulation of key signaling and metabolic enzymes. Under NR-S:M conditions, early apoptotic events and the pro-apoptotic Bcl-xS/Bcl-xL ratio were found increased. NR-S:M induced the highest retention in cellular SubG1 phase, consistent with the presence of DNA fragments from cellular apoptosis. Mitochondrial functionality, Mitochondrial ATP-linked respiration, Maximal respiration and Spare respiratory capacity, were all found blunted under NR-S:M conditions. Basal Glycolysis, Glycolytic reserve, and glycolytic capacity, together with the expression of glycogenic (PKM), gluconeogenic (PCK1 and G6PC3), and glycogenolytic enzymes (PYGL, PGM1, and G6PC3), were also negatively impacted by NR-S:M. Lastly, a TMT-proteomic approach corroborated the synchronization of liver cancer metabolic reprogramming with the activation of molecular pathways to drive a quiescent-like status of energetic-collapse and cellular death.

Conclusion: Altogether, we show that the energy-based polytherapy NR-S:M blunts cellular, metabolic and molecular plasticity of liver cancer. Notwithstanding the in vitro design of this study, it holds a promising therapeutic tool worthy of exploration for this tumor pathology.

目的:在癌症治疗中,针对葡萄糖和氧化代谢的双重干预正受到越来越多的关注。索拉非尼(Sorafenib,S)和二甲双胍(Metformin,M)这两种治疗肝癌的金标准因其线粒体抑制能力而闻名。禁食作为一种葡萄糖限制策略,也正在成为化疗的辅助手段。在此,我们探讨了营养限制联合索拉非尼:二甲双胍(NR-S:M)的抗癌反应:结果:我们的数据表明,与肝癌的侵袭性无关,禁食能协同提高索拉非尼:二甲双胍联合治疗的抗增殖效果。代谢和细胞可塑性是通过检测线粒体和糖酵解活性、细胞周期调节、细胞凋亡激活以及关键信号传导和代谢酶的调节来确定的。在 NR-S:M 条件下,发现早期凋亡事件和促凋亡 Bcl-xS/Bcl-xL 比率增加。NR-S:M 诱导的细胞 SubG1 期保留率最高,这与细胞凋亡 DNA 片段的存在一致。在 NR-S:M 条件下,线粒体功能、线粒体 ATP 链接呼吸、最大呼吸和备用呼吸能力均被削弱。基础糖酵解、糖酵解储备和糖酵解能力,以及糖原生成酶(PKM)、葡萄糖生成酶(PCK1 和 G6PC3)和糖原分解酶(PYGL、PGM1 和 G6PC3)的表达也受到 NR-S:M 的负面影响。最后,TMT-蛋白组学方法证实了肝癌代谢重编程与分子通路激活同步进行,以驱动类似静息状态的能量塌缩和细胞死亡:总之,我们的研究表明,基于能量的综合疗法 NR-S:M 可削弱肝癌的细胞、代谢和分子可塑性。尽管这项研究是体外设计的,但它是一种很有前景的治疗工具,值得对这种肿瘤病理学进行探索。
{"title":"Fasting in combination with the cocktail Sorafenib:Metformin blunts cellular plasticity and promotes liver cancer cell death via poly-metabolic exhaustion.","authors":"Juan L López-Cánovas, Beatriz Naranjo-Martínez, Alberto Diaz-Ruiz","doi":"10.1007/s13402-024-00966-2","DOIUrl":"10.1007/s13402-024-00966-2","url":null,"abstract":"<p><strong>Purpose: </strong>Dual-Interventions targeting glucose and oxidative metabolism are receiving increasing attention in cancer therapy. Sorafenib (S) and Metformin (M), two gold-standards in liver cancer, are known for their mitochondrial inhibitory capacity. Fasting, a glucose-limiting strategy, is also emerging as chemotherapy adjuvant. Herein, we explore the anti-carcinogenic response of nutrient restriction in combination with sorafenib:metformin (NR-S:M).</p><p><strong>Results: </strong>Our data demonstrates that, independently of liver cancer aggressiveness, fasting synergistically boosts the anti-proliferative effects of S:M co-treatment. Metabolic and Cellular plasticity was determined by the examination of mitochondrial and glycolytic activity, cell cycle modulation, activation of cellular apoptosis, and regulation of key signaling and metabolic enzymes. Under NR-S:M conditions, early apoptotic events and the pro-apoptotic Bcl-xS/Bcl-xL ratio were found increased. NR-S:M induced the highest retention in cellular SubG1 phase, consistent with the presence of DNA fragments from cellular apoptosis. Mitochondrial functionality, Mitochondrial ATP-linked respiration, Maximal respiration and Spare respiratory capacity, were all found blunted under NR-S:M conditions. Basal Glycolysis, Glycolytic reserve, and glycolytic capacity, together with the expression of glycogenic (PKM), gluconeogenic (PCK1 and G6PC3), and glycogenolytic enzymes (PYGL, PGM1, and G6PC3), were also negatively impacted by NR-S:M. Lastly, a TMT-proteomic approach corroborated the synchronization of liver cancer metabolic reprogramming with the activation of molecular pathways to drive a quiescent-like status of energetic-collapse and cellular death.</p><p><strong>Conclusion: </strong>Altogether, we show that the energy-based polytherapy NR-S:M blunts cellular, metabolic and molecular plasticity of liver cancer. Notwithstanding the in vitro design of this study, it holds a promising therapeutic tool worthy of exploration for this tumor pathology.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"161-182"},"PeriodicalIF":6.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850423/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141578973","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
Retraction Note: Leptin promotes the migration and invasion of breast cancer cells by upregulating ACAT2. 注:瘦素通过上调ACAT2促进乳腺癌细胞的迁移和侵袭。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-02-01 DOI: 10.1007/s13402-025-01035-y
Yunxiu Huang, Qianni Jin, Min Su, Feihu Ji, Nian Wang, Changli Zhong, Yulin Jiang, Yifeng Liu, Zhiqian Zhang, Junhong Yang, Lan Wei, Tingmei Chen, Bing Li
{"title":"Retraction Note: Leptin promotes the migration and invasion of breast cancer cells by upregulating ACAT2.","authors":"Yunxiu Huang, Qianni Jin, Min Su, Feihu Ji, Nian Wang, Changli Zhong, Yulin Jiang, Yifeng Liu, Zhiqian Zhang, Junhong Yang, Lan Wei, Tingmei Chen, Bing Li","doi":"10.1007/s13402-025-01035-y","DOIUrl":"10.1007/s13402-025-01035-y","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"265"},"PeriodicalIF":4.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12630171/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143000855","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
Pre-existing cell subpopulations in primary prostate cancer tumors display surface fingerprints of docetaxel-resistant cells. 原发性前列腺癌肿瘤中预先存在的细胞亚群显示出多西他赛耐药细胞的表面指纹。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2025-02-01 Epub Date: 2024-08-20 DOI: 10.1007/s13402-024-00982-2
Stanislav Drápela, Barbora Kvokačková, Eva Slabáková, Anna Kotrbová, Kristína Gömöryová, Radek Fedr, Daniela Kurfürstová, Martin Eliáš, Vladimír Študent, Frederika Lenčéšová, Ganji Sri Ranjani, Vendula Pospíchalová, Vítězslav Bryja, Wytske M van Weerden, Martin Puhr, Zoran Culig, Jan Bouchal, Karel Souček

Purpose: Docetaxel resistance is a significant obstacle in the treatment of prostate cancer (PCa), resulting in unfavorable patient prognoses. Intratumoral heterogeneity, often associated with epithelial-to-mesenchymal transition (EMT), has previously emerged as a phenomenon that facilitates adaptation to various stimuli, thus promoting cancer cell diversity and eventually resistance to chemotherapy, including docetaxel. Hence, understanding intratumoral heterogeneity is essential for better patient prognosis and the development of personalized treatment strategies.

Methods: To address this, we employed a high-throughput single-cell flow cytometry approach to identify a specific surface fingerprint associated with docetaxel-resistance in PCa cells and complemented it with proteomic analysis of extracellular vesicles. We further validated selected antigens using docetaxel-resistant patient-derived xenografts in vivo and probed primary PCa specimens to interrogate of their surface fingerprint.

Results: Our approaches revealed a 6-molecule surface fingerprint linked to docetaxel resistance in primary PCa specimens. We observed consistent overexpression of CD95 (FAS/APO-1), and SSEA-4 surface antigens in both in vitro and in vivo docetaxel-resistant models, which was also observed in a cell subpopulation of primary PCa tumors exhibiting EMT features. Furthermore, CD95, along with the essential enzymes involved in SSEA-4 synthesis, ST3GAL1, and ST3GAL2, displayed a significant increase in patients with PCa undergoing docetaxel-based therapy, correlating with poor survival outcomes.

Conclusion: In summary, we demonstrate that the identified 6-molecule surface fingerprint associated with docetaxel resistance pre-exists in a subpopulation of primary PCa tumors before docetaxel treatment. Thus, this fingerprint warrants further validation as a promising predictive tool for docetaxel resistance in PCa patients prior to therapy initiation.

目的:多西他赛耐药性是治疗前列腺癌(PCa)的一大障碍,导致患者预后不良。瘤内异质性通常与上皮细胞向间质转化(EMT)有关,以前曾被认为是一种有利于适应各种刺激的现象,从而促进了癌细胞的多样性,最终导致对包括多西他赛在内的化疗产生耐药性。因此,了解瘤内异质性对于改善患者预后和制定个性化治疗策略至关重要:为了解决这个问题,我们采用了一种高通量单细胞流式细胞仪方法来识别与多西他赛耐药性相关的PCa细胞特异性表面指纹,并辅以细胞外囊泡的蛋白质组学分析。我们利用体内多西他赛耐药患者衍生异种移植物进一步验证了所选抗原,并对原发性PCa标本进行了探查,以研究其表面指纹:我们的方法揭示了与原发性 PCa 标本中多西他赛耐药相关的 6 个分子表面指纹。我们在体外和体内多西他赛耐药模型中观察到了CD95(FAS/APO-1)和SSEA-4表面抗原的一致过表达,在表现出EMT特征的原发性PCa肿瘤细胞亚群中也观察到了这一现象。此外,在接受多西他赛治疗的 PCa 患者中,CD95 以及参与 SSEA-4 合成的重要酶 ST3GAL1 和 ST3GAL2 的数量显著增加,这与患者的不良生存预后有关:总之,我们证明了在多西他赛治疗前,与多西他赛耐药相关的6分子表面指纹已存在于原发性PCa肿瘤亚群中。因此,这一指纹值得进一步验证,它有望在开始治疗前成为预测 PCa 患者多西他赛耐药性的工具。
{"title":"Pre-existing cell subpopulations in primary prostate cancer tumors display surface fingerprints of docetaxel-resistant cells.","authors":"Stanislav Drápela, Barbora Kvokačková, Eva Slabáková, Anna Kotrbová, Kristína Gömöryová, Radek Fedr, Daniela Kurfürstová, Martin Eliáš, Vladimír Študent, Frederika Lenčéšová, Ganji Sri Ranjani, Vendula Pospíchalová, Vítězslav Bryja, Wytske M van Weerden, Martin Puhr, Zoran Culig, Jan Bouchal, Karel Souček","doi":"10.1007/s13402-024-00982-2","DOIUrl":"10.1007/s13402-024-00982-2","url":null,"abstract":"<p><strong>Purpose: </strong>Docetaxel resistance is a significant obstacle in the treatment of prostate cancer (PCa), resulting in unfavorable patient prognoses. Intratumoral heterogeneity, often associated with epithelial-to-mesenchymal transition (EMT), has previously emerged as a phenomenon that facilitates adaptation to various stimuli, thus promoting cancer cell diversity and eventually resistance to chemotherapy, including docetaxel. Hence, understanding intratumoral heterogeneity is essential for better patient prognosis and the development of personalized treatment strategies.</p><p><strong>Methods: </strong>To address this, we employed a high-throughput single-cell flow cytometry approach to identify a specific surface fingerprint associated with docetaxel-resistance in PCa cells and complemented it with proteomic analysis of extracellular vesicles. We further validated selected antigens using docetaxel-resistant patient-derived xenografts in vivo and probed primary PCa specimens to interrogate of their surface fingerprint.</p><p><strong>Results: </strong>Our approaches revealed a 6-molecule surface fingerprint linked to docetaxel resistance in primary PCa specimens. We observed consistent overexpression of CD95 (FAS/APO-1), and SSEA-4 surface antigens in both in vitro and in vivo docetaxel-resistant models, which was also observed in a cell subpopulation of primary PCa tumors exhibiting EMT features. Furthermore, CD95, along with the essential enzymes involved in SSEA-4 synthesis, ST3GAL1, and ST3GAL2, displayed a significant increase in patients with PCa undergoing docetaxel-based therapy, correlating with poor survival outcomes.</p><p><strong>Conclusion: </strong>In summary, we demonstrate that the identified 6-molecule surface fingerprint associated with docetaxel resistance pre-exists in a subpopulation of primary PCa tumors before docetaxel treatment. Thus, this fingerprint warrants further validation as a promising predictive tool for docetaxel resistance in PCa patients prior to therapy initiation.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"205-218"},"PeriodicalIF":6.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850551/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142003719","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
Retraction Note: Downregulation of beta-catenin in chemo-tolerant TNBC through changes in receptor and antagonist profiles of the WNT pathway: Clinical and prognostic implications. 在耐化疗TNBC中,β -连环蛋白通过WNT通路受体和拮抗剂谱的改变而下调:临床和预后意义。
IF 4.8 2区 医学 Q1 Medicine Pub Date : 2025-02-01 DOI: 10.1007/s13402-025-01036-x
Saimul Islam, Hemantika Dasgupta, Mukta Basu, Anup Roy, Neyaz Alam, Susanta Roychoudhury, Chinmay Kumar Panda
{"title":"Retraction Note: Downregulation of beta-catenin in chemo-tolerant TNBC through changes in receptor and antagonist profiles of the WNT pathway: Clinical and prognostic implications.","authors":"Saimul Islam, Hemantika Dasgupta, Mukta Basu, Anup Roy, Neyaz Alam, Susanta Roychoudhury, Chinmay Kumar Panda","doi":"10.1007/s13402-025-01036-x","DOIUrl":"10.1007/s13402-025-01036-x","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"267"},"PeriodicalIF":4.8,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12627114/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143000930","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
Next-generation BCMA-targeted chimeric antigen receptor CARTemis-1: the impact of manufacturing procedure on CAR T-cell features. 下一代 BCMA 靶向嵌合抗原受体 CARTemis-1:制造过程对 CAR T 细胞特征的影响。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2025-02-01 Epub Date: 2024-08-27 DOI: 10.1007/s13402-024-00984-0
Belén Sierro-Martínez, Virginia Escamilla-Gómez, Laura Pérez-Ortega, Beatriz Guijarro-Albaladejo, Paola Hernández-Díaz, María de la Rosa-Garrido, Maribel Lara-Chica, Alfonso Rodríguez-Gil, Juan Luis Reguera-Ortega, Luzalba Sanoja-Flores, Blanca Arribas-Arribas, Miguel Ángel Montiel-Aguilera, Gloria Carmona, Maria Jose Robles, Teresa Caballero-Velázquez, Javier Briones, Hermann Einsele, Michael Hudecek, Jose Antonio Pérez-Simón, Estefanía García-Guerrero

Purpose: CAR therapy targeting BCMA is under investigation as treatment for multiple myeloma. However, given the lack of plateau in most studies, pursuing more effective alternatives is imperative. We present the preclinical and clinical validation of a new optimized anti-BCMA CAR (CARTemis-1). In addition, we explored how the manufacturing process could impact CAR-T cell product quality and fitness.

Methods: CARTemis-1 optimizations were evaluated at the preclinical level both, in vitro and in vivo. CARTemis-1 generation was validated under GMP conditions, studying the dynamics of the immunophenotype from leukapheresis to final product. Here, we studied the impact of the manufacturing process on CAR-T cells to define optimal cell culture protocol and expansion time to increase product fitness.

Results: Two different versions of CARTemis-1 with different spacers were compared. The longer version showed increased cytotoxicity. The incorporation of the safety-gene EGFRt into the CARTemis-1 structure can be used as a monitoring marker. CARTemis-1 showed no inhibition by soluble BCMA and presents potent antitumor effects both in vitro and in vivo. Expansion with IL-2 or IL-7/IL-15 was compared, revealing greater proliferation, less differentiation, and less exhaustion with IL-7/IL-15. Three consecutive batches of CARTemis-1 were produced under GMP guidelines meeting all the required specifications. CARTemis-1 cells manufactured under GMP conditions showed increased memory subpopulations, reduced exhaustion markers and selective antitumor efficacy against MM cell lines and primary myeloma cells. The optimal release time points for obtaining the best fit product were > 6 and < 10 days (days 8-10).

Conclusions: CARTemis-1 has been rationally designed to increase antitumor efficacy, overcome sBCMA inhibition, and incorporate the expression of a safety-gene. The generation of CARTemis-1 was successfully validated under GMP standards. A phase I/II clinical trial for patients with multiple myeloma will be conducted (EuCT number 2022-503063-15-00).

目的:作为多发性骨髓瘤的治疗方法,以 BCMA 为靶点的 CAR 疗法正在接受研究。然而,由于大多数研究都缺乏高原效应,因此寻求更有效的替代疗法势在必行。我们介绍了一种新型优化抗 BCMA CAR(CARTemis-1)的临床前和临床验证。此外,我们还探讨了生产过程如何影响 CAR-T 细胞产品质量和适用性:方法:我们在体外和体内对 CARTemis-1 的临床前优化进行了评估。在 GMP 条件下验证了 CARTemis-1 的生成,研究了从白细胞分离到最终产品的免疫表型动态。在此,我们研究了生产过程对 CAR-T 细胞的影响,以确定最佳细胞培养方案和扩增时间,从而提高产品的适应性:结果:我们比较了带有不同间隔的两种不同版本的 CARTemis-1。结果:比较了带有不同间隔物的两种不同版本的CARTemis-1。在 CARTemis-1 结构中加入安全基因 EGFRt 可用作监测标记。CARTemis-1没有受到可溶性BCMA的抑制,在体外和体内都有很强的抗肿瘤作用。通过比较 IL-2 或 IL-7/IL-15 的扩增效果,发现 IL-7/IL-15 的扩增效果更好,分化更少,耗竭更少。连续三批 CARTemis-1 都是在 GMP 指导下生产的,符合所有要求的规格。在 GMP 条件下生产的 CARTemis-1 细胞显示出记忆亚群增加、衰竭标志物减少以及对 MM 细胞系和原发性骨髓瘤细胞的选择性抗肿瘤功效。获得最佳产品的最佳释放时间点大于 6 和结论:CARTemis-1 经过合理设计,提高了抗肿瘤疗效,克服了 sBCMA 抑制作用,并加入了安全基因的表达。CARTemis-1 的生产成功通过了 GMP 标准的验证。将对多发性骨髓瘤患者进行 I/II 期临床试验(EuCT 编号 2022-503063-15-00)。
{"title":"Next-generation BCMA-targeted chimeric antigen receptor CARTemis-1: the impact of manufacturing procedure on CAR T-cell features.","authors":"Belén Sierro-Martínez, Virginia Escamilla-Gómez, Laura Pérez-Ortega, Beatriz Guijarro-Albaladejo, Paola Hernández-Díaz, María de la Rosa-Garrido, Maribel Lara-Chica, Alfonso Rodríguez-Gil, Juan Luis Reguera-Ortega, Luzalba Sanoja-Flores, Blanca Arribas-Arribas, Miguel Ángel Montiel-Aguilera, Gloria Carmona, Maria Jose Robles, Teresa Caballero-Velázquez, Javier Briones, Hermann Einsele, Michael Hudecek, Jose Antonio Pérez-Simón, Estefanía García-Guerrero","doi":"10.1007/s13402-024-00984-0","DOIUrl":"10.1007/s13402-024-00984-0","url":null,"abstract":"<p><strong>Purpose: </strong>CAR therapy targeting BCMA is under investigation as treatment for multiple myeloma. However, given the lack of plateau in most studies, pursuing more effective alternatives is imperative. We present the preclinical and clinical validation of a new optimized anti-BCMA CAR (CARTemis-1). In addition, we explored how the manufacturing process could impact CAR-T cell product quality and fitness.</p><p><strong>Methods: </strong>CARTemis-1 optimizations were evaluated at the preclinical level both, in vitro and in vivo. CARTemis-1 generation was validated under GMP conditions, studying the dynamics of the immunophenotype from leukapheresis to final product. Here, we studied the impact of the manufacturing process on CAR-T cells to define optimal cell culture protocol and expansion time to increase product fitness.</p><p><strong>Results: </strong>Two different versions of CARTemis-1 with different spacers were compared. The longer version showed increased cytotoxicity. The incorporation of the safety-gene EGFRt into the CARTemis-1 structure can be used as a monitoring marker. CARTemis-1 showed no inhibition by soluble BCMA and presents potent antitumor effects both in vitro and in vivo. Expansion with IL-2 or IL-7/IL-15 was compared, revealing greater proliferation, less differentiation, and less exhaustion with IL-7/IL-15. Three consecutive batches of CARTemis-1 were produced under GMP guidelines meeting all the required specifications. CARTemis-1 cells manufactured under GMP conditions showed increased memory subpopulations, reduced exhaustion markers and selective antitumor efficacy against MM cell lines and primary myeloma cells. The optimal release time points for obtaining the best fit product were > 6 and < 10 days (days 8-10).</p><p><strong>Conclusions: </strong>CARTemis-1 has been rationally designed to increase antitumor efficacy, overcome sBCMA inhibition, and incorporate the expression of a safety-gene. The generation of CARTemis-1 was successfully validated under GMP standards. A phase I/II clinical trial for patients with multiple myeloma will be conducted (EuCT number 2022-503063-15-00).</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"219-237"},"PeriodicalIF":6.6,"publicationDate":"2025-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11850460/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142079286","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
Regulatory mechanisms of steroid hormone receptors on gene transcription through chromatin interaction and enhancer reprogramming. 类固醇激素受体通过染色质相互作用和增强子重编程对基因转录的调控机制。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-12-01 Epub Date: 2024-11-14 DOI: 10.1007/s13402-024-01011-y
Ge Sun, Chunguang Zhao, Jing Han, Shaoya Wu, Yan Chen, Jing Yao, Li Li

Regulation of steroid hormone receptors (SHRs) on transcriptional reprogramming is crucial for breast cancer progression. SHRs, including estrogen receptor (ER), androgen receptor (AR), progesterone receptor (PR), and glucocorticoid receptor (GR) play key roles in remodeling the transcriptome of breast cancer cells. However, the molecular mechanisms by which SHRs regulate chromatin landscape in enhancer regions and transcription factor interactions remain largely unknown. In this review, we summarized the regulatory effects of 3 types of SHRs (AR, PR, and GR) on gene transcription through chromatin interactions and enhancer reprogramming. Specifically, AR and PR exhibit bi-directional regulatory effects (both inhibitory and promoting) on ER-mediated gene transcription, while GR modulates the transcription of pro-proliferation genes in ER-positive breast cancer cells. In addition, we have presented four enhancer reprogramming mechanisms (transcription factor cooperation, pioneer factor binding, dynamic assisted loading, and tethering) and the multiple enhancer-promoter contact models. Based on these mechanisms and models, this review proposes that the combination of multiple therapy strategies such as agonists/antagonists of SHRs plus endocrine therapy and the adoption of the latest sequencing technologies are expected to improve the efficacy of ER positive breast cancer treatment.

类固醇激素受体(SHRs)对转录重编程的调控对乳腺癌的进展至关重要。包括雌激素受体(ER)、雄激素受体(AR)、孕酮受体(PR)和糖皮质激素受体(GR)在内的类固醇激素受体在重塑乳腺癌细胞转录组方面发挥着关键作用。然而,SHRs调控增强子区域染色质景观和转录因子相互作用的分子机制在很大程度上仍然未知。在这篇综述中,我们总结了 3 种 SHR(AR、PR 和 GR)通过染色质相互作用和增强子重编程对基因转录的调控作用。具体来说,AR和PR对ER介导的基因转录具有双向调控作用(既有抑制作用,也有促进作用),而GR则能调节ER阳性乳腺癌细胞中促增殖基因的转录。此外,我们还介绍了四种增强子重编程机制(转录因子合作、先锋因子结合、动态辅助加载和系链)以及增强子-启动子多重接触模型。基于这些机制和模型,本综述提出了多种治疗策略的组合,如SHRs的激动剂/拮抗剂加内分泌治疗,以及采用最新的测序技术,有望提高ER阳性乳腺癌的治疗效果。
{"title":"Regulatory mechanisms of steroid hormone receptors on gene transcription through chromatin interaction and enhancer reprogramming.","authors":"Ge Sun, Chunguang Zhao, Jing Han, Shaoya Wu, Yan Chen, Jing Yao, Li Li","doi":"10.1007/s13402-024-01011-y","DOIUrl":"10.1007/s13402-024-01011-y","url":null,"abstract":"<p><p>Regulation of steroid hormone receptors (SHRs) on transcriptional reprogramming is crucial for breast cancer progression. SHRs, including estrogen receptor (ER), androgen receptor (AR), progesterone receptor (PR), and glucocorticoid receptor (GR) play key roles in remodeling the transcriptome of breast cancer cells. However, the molecular mechanisms by which SHRs regulate chromatin landscape in enhancer regions and transcription factor interactions remain largely unknown. In this review, we summarized the regulatory effects of 3 types of SHRs (AR, PR, and GR) on gene transcription through chromatin interactions and enhancer reprogramming. Specifically, AR and PR exhibit bi-directional regulatory effects (both inhibitory and promoting) on ER-mediated gene transcription, while GR modulates the transcription of pro-proliferation genes in ER-positive breast cancer cells. In addition, we have presented four enhancer reprogramming mechanisms (transcription factor cooperation, pioneer factor binding, dynamic assisted loading, and tethering) and the multiple enhancer-promoter contact models. Based on these mechanisms and models, this review proposes that the combination of multiple therapy strategies such as agonists/antagonists of SHRs plus endocrine therapy and the adoption of the latest sequencing technologies are expected to improve the efficacy of ER positive breast cancer treatment.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2073-2090"},"PeriodicalIF":6.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615398","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
USP28 promotes tumor progression and glycolysis by stabilizing PKM2/Hif1-α in cholangiocarcinoma. USP28 通过稳定胆管癌中的 PKM2/Hif1-α 促进肿瘤进展和糖酵解。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-12-01 Epub Date: 2024-10-17 DOI: 10.1007/s13402-024-01002-z
Qian Qiao, Jifei Wang, Shuochen Liu, Jiang Chang, Tao Zhou, Changxian Li, Yaodong Zhang, Wangjie Jiang, Yananlan Chen, Xiao Xu, Mingyu Wu, Xiangcheng Li

Background: Ubiquitination is one of the important modification of proteins which can be reversed by deubiquitinating enzymes (DUBs). Ubiquitin specific protease 28 (USP28) belongs to the deubiquitinase family, which plays a cancer-promoting function in many types of cancers such as pancreatic cancer and breast cancer. So far, the molecular function and significance of USP 28 in cholangiocarcinoma remain unclear.

Methods: In this study, we evaluated the expression of USP28 using tissue microarray (TMA), reverse transcription polymerase chain reaction (qRT-PCR), and online databases. We investigated the effect of USP28 on the progression of CCA through in vitro and in vivo functional experiments. In addition, we explored downstream molecular pathways using Western blotting (WB), immunofluorescence (IF), and mass spectrometry techniques.

Results: Here, we found that cholangiocarcinoma tissue had higher USP 28 expression than normal bile duct tissue, and that high USP 28 levels were significantly associated with a malignant phenotype and poorer prognosis in cholangiocarcinoma patients. Both in vitro and in vivo, USP28 could mediate the deubiquitination of PKM2, thereby activating the downstream Hif1-α signaling pathway, promoting glycolysis and energy supply, and finally promoting tumor progression.

Conclusion: In summary, USP28 activated downstream Hif1-α by reducing the ubiquitination level of PKM2, furthermore, promoting the level of glycolysis in CCA cells for tumor progression.

背景:泛素化是蛋白质的重要修饰之一,可通过去泛素化酶(DUBs)逆转。泛素特异性蛋白酶28(USP28)属于去泛素化酶家族,在胰腺癌、乳腺癌等多种癌症中发挥促癌作用。迄今为止,USP 28 在胆管癌中的分子功能和意义仍不清楚:在本研究中,我们使用组织芯片(TMA)、逆转录聚合酶链反应(qRT-PCR)和在线数据库评估了 USP28 的表达。我们通过体外和体内功能实验研究了 USP28 对 CCA 进展的影响。此外,我们还利用 Western 印迹(WB)、免疫荧光(IF)和质谱技术探索了下游分子通路:结果:我们发现胆管癌组织的 USP 28 表达高于正常胆管组织,而且高 USP 28 水平与胆管癌患者的恶性表型和较差的预后显著相关。在体外和体内,USP28都能介导PKM2的去泛素化,从而激活下游Hif1-α信号通路,促进糖酵解和能量供应,最终促进肿瘤进展:综上所述,USP28通过降低PKM2的泛素化水平激活了下游的Hif1-α,进一步促进了CCA细胞的糖酵解水平,从而促进了肿瘤的进展。
{"title":"USP28 promotes tumor progression and glycolysis by stabilizing PKM2/Hif1-α in cholangiocarcinoma.","authors":"Qian Qiao, Jifei Wang, Shuochen Liu, Jiang Chang, Tao Zhou, Changxian Li, Yaodong Zhang, Wangjie Jiang, Yananlan Chen, Xiao Xu, Mingyu Wu, Xiangcheng Li","doi":"10.1007/s13402-024-01002-z","DOIUrl":"10.1007/s13402-024-01002-z","url":null,"abstract":"<p><strong>Background: </strong>Ubiquitination is one of the important modification of proteins which can be reversed by deubiquitinating enzymes (DUBs). Ubiquitin specific protease 28 (USP28) belongs to the deubiquitinase family, which plays a cancer-promoting function in many types of cancers such as pancreatic cancer and breast cancer. So far, the molecular function and significance of USP 28 in cholangiocarcinoma remain unclear.</p><p><strong>Methods: </strong>In this study, we evaluated the expression of USP28 using tissue microarray (TMA), reverse transcription polymerase chain reaction (qRT-PCR), and online databases. We investigated the effect of USP28 on the progression of CCA through in vitro and in vivo functional experiments. In addition, we explored downstream molecular pathways using Western blotting (WB), immunofluorescence (IF), and mass spectrometry techniques.</p><p><strong>Results: </strong>Here, we found that cholangiocarcinoma tissue had higher USP 28 expression than normal bile duct tissue, and that high USP 28 levels were significantly associated with a malignant phenotype and poorer prognosis in cholangiocarcinoma patients. Both in vitro and in vivo, USP28 could mediate the deubiquitination of PKM2, thereby activating the downstream Hif1-α signaling pathway, promoting glycolysis and energy supply, and finally promoting tumor progression.</p><p><strong>Conclusion: </strong>In summary, USP28 activated downstream Hif1-α by reducing the ubiquitination level of PKM2, furthermore, promoting the level of glycolysis in CCA cells for tumor progression.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"2217-2231"},"PeriodicalIF":6.6,"publicationDate":"2024-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142459017","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
Cellular Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1