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 : 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":"https://doi.org/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":""},"PeriodicalIF":6.6,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","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":"","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 : 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":"https://doi.org/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":""},"PeriodicalIF":6.6,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","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":"","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 : 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":"https://doi.org/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":""},"PeriodicalIF":6.6,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","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":"","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 : 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":"https://doi.org/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":""},"PeriodicalIF":6.6,"publicationDate":"2024-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","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":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Omics-based molecular classifications empowering in precision oncology. 基于组学的分子分类为精准肿瘤学赋能。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-06-01 Epub Date: 2024-01-31 DOI: 10.1007/s13402-023-00912-8
Zhaokai Zhou, Ting Lin, Shuang Chen, Ge Zhang, Yudi Xu, Haijiao Zou, Aoyang Zhou, Yuyuan Zhang, Siyuan Weng, Xinwei Han, Zaoqu Liu

Background: In the past decades, cancer enigmatical heterogeneity at distinct expression levels could interpret disparities in therapeutic response and prognosis. It built hindrances to precision medicine, a tactic to tailor customized treatment informed by the tumors' molecular profile. Single-omics analysis dissected the biological features associated with carcinogenesis to some extent but still failed to revolutionize cancer treatment as expected. Integrated omics analysis incorporated tumor biological networks from diverse layers and deciphered a holistic overview of cancer behaviors, yielding precise molecular classification to facilitate the evolution and refinement of precision medicine.

Conclusion: This review outlined the biomarkers at multiple expression layers to tutor molecular classification and pinpoint tumor diagnosis, and explored the paradigm shift in precision therapy: from single- to multi-omics-based subtyping to optimize therapeutic regimens. Ultimately, we firmly believe that by parsing molecular characteristics, omics-based typing will be a powerful assistant for precision oncology.

背景:在过去的几十年里,癌症在不同表达水平上的神秘异质性可以解释治疗反应和预后的差异。它阻碍了精准医疗的发展,而精准医疗是一种根据肿瘤分子特征量身定制治疗方案的策略。单一组学分析在一定程度上剖析了与癌变相关的生物学特征,但仍未能如预期那样彻底改变癌症治疗。综合组学分析从不同层面整合了肿瘤生物网络,解读了癌症行为的整体概况,得出了精确的分子分类,促进了精准医疗的发展和完善:本综述概述了多个表达层的生物标志物,以指导分子分类和精确诊断肿瘤,并探讨了精准治疗的范式转变:从基于单一组学的亚型分析到基于多组学的亚型分析,以优化治疗方案。最终,我们坚信,通过解析分子特征,基于组学的分型将成为精准肿瘤学的有力助手。
{"title":"Omics-based molecular classifications empowering in precision oncology.","authors":"Zhaokai Zhou, Ting Lin, Shuang Chen, Ge Zhang, Yudi Xu, Haijiao Zou, Aoyang Zhou, Yuyuan Zhang, Siyuan Weng, Xinwei Han, Zaoqu Liu","doi":"10.1007/s13402-023-00912-8","DOIUrl":"10.1007/s13402-023-00912-8","url":null,"abstract":"<p><strong>Background: </strong>In the past decades, cancer enigmatical heterogeneity at distinct expression levels could interpret disparities in therapeutic response and prognosis. It built hindrances to precision medicine, a tactic to tailor customized treatment informed by the tumors' molecular profile. Single-omics analysis dissected the biological features associated with carcinogenesis to some extent but still failed to revolutionize cancer treatment as expected. Integrated omics analysis incorporated tumor biological networks from diverse layers and deciphered a holistic overview of cancer behaviors, yielding precise molecular classification to facilitate the evolution and refinement of precision medicine.</p><p><strong>Conclusion: </strong>This review outlined the biomarkers at multiple expression layers to tutor molecular classification and pinpoint tumor diagnosis, and explored the paradigm shift in precision therapy: from single- to multi-omics-based subtyping to optimize therapeutic regimens. Ultimately, we firmly believe that by parsing molecular characteristics, omics-based typing will be a powerful assistant for precision oncology.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"759-777"},"PeriodicalIF":6.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139641695","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
E3 ligase SOCS3 regulates NOD2 expression by ubiquitin proteasome system in lung cancer progression. E3连接酶SOCS3通过泛素-蛋白酶体系统调节NOD2在癌症进展中的表达。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-06-01 Epub Date: 2023-11-01 DOI: 10.1007/s13402-023-00896-5
In-Ho Jeong, Jae Kwang Yun, Jun-O Jin, Jeong Hee Hong, Ji Yeon Lee, Geun Dong Lee, Peter Chang-Whan Lee

Purpose: Despite lung cancer is one of the leading causes of cancer-related deaths, it remains hard to discover effective diagnostic and therapeutic approaches. Moreover, the five-year survival rate is relatively lower than other tumors. So urgent needs for finding a new theranostic target to treat lung cancer effectively. This study aims to present SOCS3 and NOD2 proteins as novel targets for diagnosis and therapy.

Methods: We first confirmed SOCS3 expression level in patients' tissues. Then, we applied knockdown and overexpression of SOCS3 on lung cancer cell lines and performed proliferation, migration, and invasion assay. After that, we found NOD2 is a target of SOCS3 and introduced overexpression of NOD2 to A549 for verifying reduced tumorigenicity of lung cancer cells.

Results: We identified protein expression level of SOCS3 was frequently higher in tumor tissues than adjacent normal tissues. Truly, overexpression of SOCS3 promoted proliferation, migration, and invasion capacity of lung cancer cells. We found that SOCS3 interacts with NOD2 and SOCS3 ubiquitinates NOD2 directly. Furthermore, lung cancer tissues with higher SOCS3 expression showed lower NOD2 expression. We confirmed overexpression of NOD2 leads to suppressed tumorigenicity of lung cancer cells, and these effects occurred through MAPK pathway.

Conclusion: Collectively, our work reveals novel roles of SOCS3 in lung tumorigenesis and proposes SOCS3 as a promising biomarker candidate for therapeutic and diagnostic target for lung cancer.

目的:尽管癌症是癌症相关死亡的主要原因之一,但仍很难发现有效的诊断和治疗方法。此外,五年生存率相对低于其他肿瘤。因此迫切需要寻找一种新的治疗靶点来有效治疗癌症。本研究旨在提出SOCS3和NOD2蛋白作为诊断和治疗的新靶点。方法:我们首先确认了SOCS3在患者组织中的表达水平。然后,我们在癌症细胞系上应用SOCS3的敲低和过表达,并进行增殖、迁移和侵袭测定。之后,我们发现NOD2是SOCS3的靶点,并将NOD2过度表达引入A549,以验证癌症细胞的致瘤性降低。结果:我们发现SOCS3在肿瘤组织中的蛋白表达水平通常高于邻近的正常组织。确实,SOCS3的过度表达促进了癌症细胞的增殖、迁移和侵袭能力。我们发现SOCS3与NOD2相互作用,并且SOCS3直接泛素化NOD2。此外,具有较高SOCS3表达的癌症组织表现出较低的NOD2表达。我们证实NOD2的过度表达可抑制癌症细胞的致瘤性,这些作用是通过MAPK途径发生的。结论:总之,我们的工作揭示了SOCS3在肺肿瘤发生中的新作用,并提出SOCS3是一种有前途的候选生物标志物,可作为癌症的治疗和诊断靶点。
{"title":"E3 ligase SOCS3 regulates NOD2 expression by ubiquitin proteasome system in lung cancer progression.","authors":"In-Ho Jeong, Jae Kwang Yun, Jun-O Jin, Jeong Hee Hong, Ji Yeon Lee, Geun Dong Lee, Peter Chang-Whan Lee","doi":"10.1007/s13402-023-00896-5","DOIUrl":"10.1007/s13402-023-00896-5","url":null,"abstract":"<p><strong>Purpose: </strong>Despite lung cancer is one of the leading causes of cancer-related deaths, it remains hard to discover effective diagnostic and therapeutic approaches. Moreover, the five-year survival rate is relatively lower than other tumors. So urgent needs for finding a new theranostic target to treat lung cancer effectively. This study aims to present SOCS3 and NOD2 proteins as novel targets for diagnosis and therapy.</p><p><strong>Methods: </strong>We first confirmed SOCS3 expression level in patients' tissues. Then, we applied knockdown and overexpression of SOCS3 on lung cancer cell lines and performed proliferation, migration, and invasion assay. After that, we found NOD2 is a target of SOCS3 and introduced overexpression of NOD2 to A549 for verifying reduced tumorigenicity of lung cancer cells.</p><p><strong>Results: </strong>We identified protein expression level of SOCS3 was frequently higher in tumor tissues than adjacent normal tissues. Truly, overexpression of SOCS3 promoted proliferation, migration, and invasion capacity of lung cancer cells. We found that SOCS3 interacts with NOD2 and SOCS3 ubiquitinates NOD2 directly. Furthermore, lung cancer tissues with higher SOCS3 expression showed lower NOD2 expression. We confirmed overexpression of NOD2 leads to suppressed tumorigenicity of lung cancer cells, and these effects occurred through MAPK pathway.</p><p><strong>Conclusion: </strong>Collectively, our work reveals novel roles of SOCS3 in lung tumorigenesis and proposes SOCS3 as a promising biomarker candidate for therapeutic and diagnostic target for lung cancer.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"819-832"},"PeriodicalIF":6.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71421047","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
CHIP promotes CAD ubiquitination and degradation to suppress the proliferation and colony formation of glioblastoma cells. CHIP促进CAD泛素化和降解,抑制胶质母细胞瘤细胞的增殖和集落形成。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-06-01 Epub Date: 2023-11-20 DOI: 10.1007/s13402-023-00899-2
Guanya Li, Kai Xiao, Yinan Li, Jianfang Gao, Shanping He, Tingting Li

Purpose: Cancer cells are characterized as the uncontrolled proliferation, which demands high levels of nucleotides that are building blocks for DNA synthesis and replication. CAD (carbamoyl-phosphate synthetase 2, aspartate transcarbamylase and dihydroorotase) is a trifunctional enzyme that initiates the de novo pyrimidine synthesis, which is normally enhanced in cancer cells to preserve the pyrimidine pool for cell division. Glioma, representing most brain cancer, is highly addicted to nucleotides like pyrimidine to sustain the abnormal growth and proliferation of cells. CAD is previously reported to be dysregulated in glioma, but the underlying mechanism remains unclear.

Methods: The expression of CAD and CHIP (carboxyl terminus of Hsc70-interacting protein) protein in normal brain cells and three glioblastoma (GBM) cell lines were measured by immunoblots. Lentiviruses-mediated expression of target proteins or shRNAs were used to specifically overexpress or knock down CAD and CHIP. Cell counting, colony formation, apoptosis and cell cycle assays were used to assess the roles of CAD and CHIP in GBM cell proliferation and survival. Co-immunoprecipitation and ubiquitination assays were used to examine the interaction of CHIP with CAD and the ubiquitination of CAD. The correlation of CAD and CHIP expression with GBM patients' survival was obtained by analyzing the GlioVis database.

Results: In this study, we showed that the expression of CAD was upregulated in glioma, which was positively correlated with the tumor grade and survival of glioma patients. Knockdown of CAD robustly inhibited the cell proliferation and colony formation of GBM cells, indicating the essential role of CAD in the pathogenesis of GBM. Mechanistically, we firstly identified that CAD was modified by the K29-linked polyubiquitination, which was mediated by the E3 ubiquitin ligase CHIP. By interacting with and ubiquitinating CAD, CHIP enhanced its proteasomal and lysosomal degradation, which accounted for the anti-proliferative role of CHIP in GBM cells. To sustain the expression of CAD, CHIP is significantly downregulated, which is correlated with the poor prognosis and survival of GBM patients. Notably, the low level of CHIP and high level of CAD overall predict the short survival of GBM patients.

Conclusion: Altogether, these results illustrated the essential role of CAD in GBM and revealed a novel therapeutic strategy for CAD-positive and CHIP-negative cancer.

目的:癌细胞的特点是不受控制的增殖,这需要高水平的核苷酸,核苷酸是DNA合成和复制的基础。CAD(氨甲酰磷酸合成酶2,天冬氨酸转氨基甲酰基酶和二氢化羧酶)是一种三功能酶,可启动新的嘧啶合成,通常在癌细胞中增强,以保存用于细胞分裂的嘧啶池。神经胶质瘤是大多数脑癌的代表,它高度依赖嘧啶等核苷酸来维持细胞的异常生长和增殖。先前有报道称,CAD在胶质瘤中失调,但其潜在机制尚不清楚。方法:采用免疫印迹法检测正常脑细胞和3种胶质母细胞瘤(GBM)细胞系中CAD和CHIP蛋白的表达。慢病毒介导的靶蛋白或shrna表达被用来特异性地过表达或敲低CAD和CHIP。通过细胞计数、集落形成、细胞凋亡和细胞周期测定来评估CAD和CHIP在GBM细胞增殖和存活中的作用。采用免疫共沉淀法和泛素化法检测CHIP与CAD的相互作用和CAD的泛素化。通过分析GlioVis数据库获得CAD和CHIP表达与GBM患者生存的相关性。结果:在本研究中,我们发现CAD在胶质瘤中表达上调,与胶质瘤患者的肿瘤分级和生存率呈正相关。敲低CAD可显著抑制GBM细胞的增殖和集落形成,提示CAD在GBM发病过程中发挥重要作用。在机制上,我们首先发现CAD被k29连接的多泛素化修饰,这是由E3泛素连接酶CHIP介导的。CHIP通过与CAD相互作用并使其泛素化,增强了其蛋白酶体和溶酶体的降解,这解释了CHIP在GBM细胞中的抗增殖作用。为了维持CAD的表达,CHIP显著下调,这与GBM患者预后不良和生存相关。值得注意的是,低CHIP水平和高CAD水平总体上预示着GBM患者的短生存期。结论:总之,这些结果说明了CAD在GBM中的重要作用,并为CAD阳性和chip阴性的癌症提供了新的治疗策略。
{"title":"CHIP promotes CAD ubiquitination and degradation to suppress the proliferation and colony formation of glioblastoma cells.","authors":"Guanya Li, Kai Xiao, Yinan Li, Jianfang Gao, Shanping He, Tingting Li","doi":"10.1007/s13402-023-00899-2","DOIUrl":"10.1007/s13402-023-00899-2","url":null,"abstract":"<p><strong>Purpose: </strong>Cancer cells are characterized as the uncontrolled proliferation, which demands high levels of nucleotides that are building blocks for DNA synthesis and replication. CAD (carbamoyl-phosphate synthetase 2, aspartate transcarbamylase and dihydroorotase) is a trifunctional enzyme that initiates the de novo pyrimidine synthesis, which is normally enhanced in cancer cells to preserve the pyrimidine pool for cell division. Glioma, representing most brain cancer, is highly addicted to nucleotides like pyrimidine to sustain the abnormal growth and proliferation of cells. CAD is previously reported to be dysregulated in glioma, but the underlying mechanism remains unclear.</p><p><strong>Methods: </strong>The expression of CAD and CHIP (carboxyl terminus of Hsc70-interacting protein) protein in normal brain cells and three glioblastoma (GBM) cell lines were measured by immunoblots. Lentiviruses-mediated expression of target proteins or shRNAs were used to specifically overexpress or knock down CAD and CHIP. Cell counting, colony formation, apoptosis and cell cycle assays were used to assess the roles of CAD and CHIP in GBM cell proliferation and survival. Co-immunoprecipitation and ubiquitination assays were used to examine the interaction of CHIP with CAD and the ubiquitination of CAD. The correlation of CAD and CHIP expression with GBM patients' survival was obtained by analyzing the GlioVis database.</p><p><strong>Results: </strong>In this study, we showed that the expression of CAD was upregulated in glioma, which was positively correlated with the tumor grade and survival of glioma patients. Knockdown of CAD robustly inhibited the cell proliferation and colony formation of GBM cells, indicating the essential role of CAD in the pathogenesis of GBM. Mechanistically, we firstly identified that CAD was modified by the K29-linked polyubiquitination, which was mediated by the E3 ubiquitin ligase CHIP. By interacting with and ubiquitinating CAD, CHIP enhanced its proteasomal and lysosomal degradation, which accounted for the anti-proliferative role of CHIP in GBM cells. To sustain the expression of CAD, CHIP is significantly downregulated, which is correlated with the poor prognosis and survival of GBM patients. Notably, the low level of CHIP and high level of CAD overall predict the short survival of GBM patients.</p><p><strong>Conclusion: </strong>Altogether, these results illustrated the essential role of CAD in GBM and revealed a novel therapeutic strategy for CAD-positive and CHIP-negative cancer.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"851-865"},"PeriodicalIF":6.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138046213","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
HPV16 E6/E7-mediated regulation of PiwiL1 expression induces tumorigenesis in cervical cancer cells. hpv16e6 / e7介导的PiwiL1表达调控诱导宫颈癌细胞的肿瘤发生
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-06-01 Epub Date: 2023-12-01 DOI: 10.1007/s13402-023-00904-8
Midhunaraj Kunnummal, Pooja Sherly Raveendran, Budhaditya Basu, Sheri Vidya Rani, Riya Ann Paul, Krithiga Kuppusamy, Mary Angelin, Joby Issac, Jackson James, Ani V Das

Purpose: PiwiL1 has been reported to be over-expressed in many cancers. However, the molecular mechanism by which these proteins contribute to tumorigenesis and their regulation in cancer cells is still unclear. We intend to understand the role of PiwiL1 in tumorigenesis and also its regulation in cervical cells.

Methods: We studied the effect of loss of PiwiL1 function on tumor properties of cervical cancer cells in vitro and in vivo. Also we have looked into the effect of PiwiL1 overexpression in the malignant transformation of normal cells both in vitro and in vivo. Further RNA-seq and RIP-seq analyses were done to get insight of the direct and indirect targets of PiwiL1 in the cervical cancer cells.

Results: Here, we report that PiwiL1 is not only over-expressed, but also play a major role in tumor induction and progression. Abolition of PiwiL1 in CaSki cells led to a decrease in the tumor-associated properties, whereas, its upregulation conferred malignant transformation of normal HaCaT cells. Our study delineates a new link between HPV oncogenes, E6 and E7 with PiwiL1. p53 and E2F1 directly bind and differentially regulate PiwiL1 promoter in a context-dependant manner. Further, RNA-seq together with RIP-RNA-seq suggested a strong and direct role for PiwiL1 in promoting metastasis in cervical cancer cells.

Conclusion: Our study demonstrates that PiwiL1 act as an oncogene in cervical cancer by inducing tumor-associated properties and EMT pathway. The finding that HPV oncogenes, E6/E7 can positively regulate PiwiL1 suggests a possible mechanism behind HPV-mediated tumorigenesis in cervical cancer.

目的:据报道,PiwiL1在许多癌症中过表达。然而,这些蛋白参与肿瘤发生及其在癌细胞中的调控的分子机制尚不清楚。我们打算了解PiwiL1在肿瘤发生中的作用及其在宫颈细胞中的调节作用。方法:体外和体内研究PiwiL1功能缺失对宫颈癌细胞肿瘤特性的影响。此外,我们还在体外和体内研究了PiwiL1过表达对正常细胞恶性转化的影响。进一步进行RNA-seq和RIP-seq分析,以深入了解PiwiL1在宫颈癌细胞中的直接和间接靶点。结果:在这里,我们报道了PiwiL1不仅过表达,而且在肿瘤诱导和进展中发挥重要作用。CaSki细胞中PiwiL1的缺失导致肿瘤相关特性的降低,而其上调则导致正常HaCaT细胞的恶性转化。我们的研究描述了HPV癌基因E6和E7与PiwiL1之间的新联系。p53和E2F1以上下文依赖的方式直接结合并差异调节PiwiL1启动子。此外,RNA-seq和RIP-RNA-seq表明,PiwiL1在促进宫颈癌细胞转移中具有强烈而直接的作用。结论:我们的研究表明PiwiL1通过诱导肿瘤相关特性和EMT通路在宫颈癌中发挥致癌基因的作用。HPV致癌基因E6/E7可以正向调节PiwiL1,这一发现提示HPV介导宫颈癌肿瘤发生的可能机制。
{"title":"HPV16 E6/E7-mediated regulation of PiwiL1 expression induces tumorigenesis in cervical cancer cells.","authors":"Midhunaraj Kunnummal, Pooja Sherly Raveendran, Budhaditya Basu, Sheri Vidya Rani, Riya Ann Paul, Krithiga Kuppusamy, Mary Angelin, Joby Issac, Jackson James, Ani V Das","doi":"10.1007/s13402-023-00904-8","DOIUrl":"10.1007/s13402-023-00904-8","url":null,"abstract":"<p><strong>Purpose: </strong>PiwiL1 has been reported to be over-expressed in many cancers. However, the molecular mechanism by which these proteins contribute to tumorigenesis and their regulation in cancer cells is still unclear. We intend to understand the role of PiwiL1 in tumorigenesis and also its regulation in cervical cells.</p><p><strong>Methods: </strong>We studied the effect of loss of PiwiL1 function on tumor properties of cervical cancer cells in vitro and in vivo. Also we have looked into the effect of PiwiL1 overexpression in the malignant transformation of normal cells both in vitro and in vivo. Further RNA-seq and RIP-seq analyses were done to get insight of the direct and indirect targets of PiwiL1 in the cervical cancer cells.</p><p><strong>Results: </strong>Here, we report that PiwiL1 is not only over-expressed, but also play a major role in tumor induction and progression. Abolition of PiwiL1 in CaSki cells led to a decrease in the tumor-associated properties, whereas, its upregulation conferred malignant transformation of normal HaCaT cells. Our study delineates a new link between HPV oncogenes, E6 and E7 with PiwiL1. p53 and E2F1 directly bind and differentially regulate PiwiL1 promoter in a context-dependant manner. Further, RNA-seq together with RIP-RNA-seq suggested a strong and direct role for PiwiL1 in promoting metastasis in cervical cancer cells.</p><p><strong>Conclusion: </strong>Our study demonstrates that PiwiL1 act as an oncogene in cervical cancer by inducing tumor-associated properties and EMT pathway. The finding that HPV oncogenes, E6/E7 can positively regulate PiwiL1 suggests a possible mechanism behind HPV-mediated tumorigenesis in cervical cancer.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"917-937"},"PeriodicalIF":6.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138458175","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
The development of in vitro organotypic 3D vulvar models to study tumor-stroma interaction and drug efficacy. 建立体外器官型3D外阴模型,研究肿瘤-基质相互作用及药物疗效。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-06-01 Epub Date: 2023-12-07 DOI: 10.1007/s13402-023-00902-w
Shidi Wu, Bertine W Huisman, Marion H Rietveld, Robert Rissmann, Maarten H Vermeer, Mariette I E van Poelgeest, Abdoelwaheb El Ghalbzouri

Background: Vulvar squamous cell carcinoma (VSCC) is a rare disease with a poor prognosis. To date, there's no proper in vitro modeling system for VSCC to study its pathogenesis or for drug evaluation.

Methods: We established healthy vulvar (HV)- and VSCC-like 3D full thickness models (FTMs) to observe the tumor-stroma interaction and their applicability for chemotherapeutic efficacy examination. VSCC-FTMs were developed by seeding VSCC tumor cell lines (A431 and HTB117) onto dermal matrices harboring two NF subtypes namely papillary fibroblasts (PFs) and reticular fibroblasts (RFs), or cancer-associated fibroblasts (CAFs) while HV-FTMs were constructed with primary keratinocytes and fibroblasts isolated from HV tissues.

Results: HV-FTMs highly resembled HV tissues in terms of epidermal morphogenesis, basement membrane formation and collagen deposition. When the dermal compartment shifted from PFs to RFs or CAFs in VSCC-FTMs, tumor cells demonstrated more proliferation, EMT induction and stemness. In contrast to PFs, RFs started to lose their phenotype and express robust CAF-markers α-SMA and COL11A1 under tumor cell signaling induction, indicating a favored 'RF-to-CAF' transition in VSCC tumor microenvironment (TME). Additionally, chemotherapeutic treatment with carboplatin and paclitaxel resulted in a significant reduction in tumor-load and invasion in VSCC-FTMs.

Conclusion: We successfully developed in vitro 3D vulvar models mimicking both healthy and tumorous conditions which serve as a promising tool for vulvar drug screening programs. Moreover, healthy fibroblasts demonstrate heterogeneity in terms of CAF-activation in VSCC TME which brings insights in the future development of novel CAF-based therapeutic strategies in VSCC.

背景:外阴鳞状细胞癌(VSCC)是一种罕见的疾病,预后较差。到目前为止,还没有合适的体外造模系统来研究VSCC的发病机制或进行药物评价。方法:建立健康外阴(HV)样和vscc样三维全层模型(FTMs),观察肿瘤与间质相互作用及其在化疗疗效检验中的适用性。VSCC- ftms是通过将VSCC肿瘤细胞系(A431和HTB117)播种到含有两种NF亚型(乳头状成纤维细胞(PFs)和网状成纤维细胞(RFs)或癌症相关成纤维细胞(CAFs)的真皮基质上培育而成的,而HV- ftms是用从HV组织中分离的原代角质形成细胞和成纤维细胞构建的。结果:HV- ftms在表皮形态发生、基底膜形成和胶原沉积等方面与HV组织高度相似。在VSCC-FTMs中,当真皮隔室从PFs转移到RFs或CAFs时,肿瘤细胞表现出更多的增殖、EMT诱导和干细胞性。与PFs相比,在肿瘤细胞信号传导诱导下,RFs开始失去其表型并表达强大的caf标记α-SMA和COL11A1,这表明VSCC肿瘤微环境(TME)中更倾向于“rf到caf”的转变。此外,卡铂和紫杉醇化疗可显著降低VSCC-FTMs的肿瘤负荷和侵袭。结论:我们成功地建立了模拟健康和肿瘤情况的体外3D外阴模型,为外阴药物筛选提供了一个有前途的工具。此外,健康成纤维细胞在VSCC TME中cafa活化方面表现出异质性,这为未来开发新的基于cafa的VSCC治疗策略提供了见解。
{"title":"The development of in vitro organotypic 3D vulvar models to study tumor-stroma interaction and drug efficacy.","authors":"Shidi Wu, Bertine W Huisman, Marion H Rietveld, Robert Rissmann, Maarten H Vermeer, Mariette I E van Poelgeest, Abdoelwaheb El Ghalbzouri","doi":"10.1007/s13402-023-00902-w","DOIUrl":"10.1007/s13402-023-00902-w","url":null,"abstract":"<p><strong>Background: </strong>Vulvar squamous cell carcinoma (VSCC) is a rare disease with a poor prognosis. To date, there's no proper in vitro modeling system for VSCC to study its pathogenesis or for drug evaluation.</p><p><strong>Methods: </strong>We established healthy vulvar (HV)- and VSCC-like 3D full thickness models (FTMs) to observe the tumor-stroma interaction and their applicability for chemotherapeutic efficacy examination. VSCC-FTMs were developed by seeding VSCC tumor cell lines (A431 and HTB117) onto dermal matrices harboring two NF subtypes namely papillary fibroblasts (PFs) and reticular fibroblasts (RFs), or cancer-associated fibroblasts (CAFs) while HV-FTMs were constructed with primary keratinocytes and fibroblasts isolated from HV tissues.</p><p><strong>Results: </strong>HV-FTMs highly resembled HV tissues in terms of epidermal morphogenesis, basement membrane formation and collagen deposition. When the dermal compartment shifted from PFs to RFs or CAFs in VSCC-FTMs, tumor cells demonstrated more proliferation, EMT induction and stemness. In contrast to PFs, RFs started to lose their phenotype and express robust CAF-markers α-SMA and COL11A1 under tumor cell signaling induction, indicating a favored 'RF-to-CAF' transition in VSCC tumor microenvironment (TME). Additionally, chemotherapeutic treatment with carboplatin and paclitaxel resulted in a significant reduction in tumor-load and invasion in VSCC-FTMs.</p><p><strong>Conclusion: </strong>We successfully developed in vitro 3D vulvar models mimicking both healthy and tumorous conditions which serve as a promising tool for vulvar drug screening programs. Moreover, healthy fibroblasts demonstrate heterogeneity in terms of CAF-activation in VSCC TME which brings insights in the future development of novel CAF-based therapeutic strategies in VSCC.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":" ","pages":"883-896"},"PeriodicalIF":6.6,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138497873","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
Targeting Dectin-1 and or VISTA enhances anti-tumor immunity in melanoma but not colorectal cancer model. 靶向 Dectin-1 和或 VISTA 能增强黑色素瘤模型的抗肿瘤免疫力,但不能增强结直肠癌模型的抗肿瘤免疫力。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-04-26 DOI: 10.1007/s13402-024-00950-w
Siavash Mashhouri, Amirhossein Rahmati, Ako Azimi, Roy A Fava, Ismail Hassan Ismail, John Walker, Shokrollah Elahi
{"title":"Targeting Dectin-1 and or VISTA enhances anti-tumor immunity in melanoma but not colorectal cancer model.","authors":"Siavash Mashhouri, Amirhossein Rahmati, Ako Azimi, Roy A Fava, Ismail Hassan Ismail, John Walker, Shokrollah Elahi","doi":"10.1007/s13402-024-00950-w","DOIUrl":"https://doi.org/10.1007/s13402-024-00950-w","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":"11 8","pages":""},"PeriodicalIF":6.6,"publicationDate":"2024-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140652955","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: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