首页 > 最新文献

Oncology Research最新文献

英文 中文
Hsa-miR-214-3p inhibits breast cancer cell growth and improves the tumor immune microenvironment by downregulating B7H3. Hsa-miR-214-3p通过下调B7H3抑制乳腺癌细胞生长,改善肿瘤免疫微环境。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.057472
Yan Lu, Kang Wang, Yuanhong Peng, Meng Chen, Lin Zhong, Luji Huang, F U Cheng, Xindan Sheng, Xin Yang, Manzhao Ouyang, George A Calin, Zhiwei He

Background: Immune checkpoint inhibitors play an important role in the treatment of solid tumors, but the currently used immune checkpoint inhibitors targeting programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte antigen-4 (CTLA-4) show limited clinical efficacy in many breast cancers. B7H3 has been widely reported as an immunosuppressive molecule, but its immunological function in breast cancer patients remains unclear.

Methods: We analyzed the expression of B7H3 in breast cancer samples using data from the Cancer Genome Atlas Program (TCGA) and the Gene Expression Omnibus (GEO) databases. MicroRNAs were selected using the TarBase, miRTarBase, and miRBase databases. The regulatory role of the microRNA hsa-miR-214-3p on B7H3 was investigated through dual-luciferase reporter assays, which identified the specific action sites of interaction. The expression levels of B7H3 and hsa-miR-214-3p in human breast cancer tissues and adjacent normal tissues were quantified using Western blotting and quantitative PCR (qPCR). In vitro experiments were performed to observe the effects of modulating the expression of B7H3 or hsa-miR-214-3p on breast cancer cell proliferation and apoptosis. Additionally, the regulatory impact of hsa-miR-214-3p on B7H3 was examined. Enzyme-linked immunosorbent assays (ELISA) and flow cytometry were employed to assess the effects of co-cultured breast cancer cells and normal human peripheral blood mononuclear cells (PBMCs) on immune cells and associated cytokines.

Results: In breast cancer tissues, the expression level of B7H3 is inversely correlated with that of hsa-miR-214-3p, as well as with the regulatory effects on breast cancercell behavior. Hsa-miR-214-3p was found to inhibit breast cancer cell growth by downregulating B7H3. Importantly, our research identified, for the first time, two binding sites for hsa-miR-214-3p on the 3' UTR of B7H3, both of which exert similar effects independently. Co-culture experiments revealed that hsa-miR-214-3p obstructs the suppressive function of B7H3 on CD8+ T cells and natural killer cells.

Conclusions: This study confirms the existence of two hsa-miR-214-3p binding sites on the 3' UTR of B7H3, reinforcing the role of hsa-miR-214-3p as a regulatory factor for B7H3. In breast cancer, hsa-miR-214-3p reduces tumor cell proliferation and enhances the tumor immune microenvironment by downregulating B7H3. These findings suggest new potential targets for the clinical treatment of breast cancer.

背景:免疫检查点抑制剂在实体肿瘤的治疗中发挥着重要作用,但目前使用的针对程序性细胞死亡-1 (PD-1)、程序性细胞死亡配体-1 (PD-L1)和细胞毒性t淋巴细胞抗原-4 (CTLA-4)的免疫检查点抑制剂在许多乳腺癌中的临床疗效有限。B7H3作为一种免疫抑制分子已被广泛报道,但其在乳腺癌患者中的免疫功能尚不清楚。方法:利用癌症基因组图谱计划(TCGA)和基因表达Omnibus (GEO)数据库的数据分析乳腺癌样本中B7H3的表达。使用TarBase、miRTarBase和miRBase数据库选择microrna。通过双荧光素酶报告基因试验研究了microRNA hsa-miR-214-3p对B7H3的调控作用,确定了相互作用的具体作用位点。采用Western blotting和定量PCR (qPCR)技术,定量检测B7H3和hsa-miR-214-3p在人乳腺癌组织及癌旁正常组织中的表达水平。通过体外实验观察调节B7H3或hsa-miR-214-3p表达对乳腺癌细胞增殖和凋亡的影响。此外,我们还检测了hsa-miR-214-3p对B7H3的调控作用。采用酶联免疫吸附法(ELISA)和流式细胞术研究乳腺癌细胞与正常人外周血单个核细胞(PBMCs)共培养对免疫细胞及相关细胞因子的影响。结果:在乳腺癌组织中,B7H3的表达水平与hsa-miR-214-3p的表达水平呈负相关,并与乳腺癌细胞行为的调节作用呈负相关。发现Hsa-miR-214-3p通过下调B7H3抑制乳腺癌细胞生长。重要的是,我们的研究首次在B7H3的3' UTR上发现了hsa-miR-214-3p的两个结合位点,这两个位点各自独立发挥相似的作用。共培养实验发现,hsa-miR-214-3p阻断了B7H3对CD8+ T细胞和自然杀伤细胞的抑制功能。结论:本研究证实了B7H3的3′UTR上存在两个hsa-miR-214-3p结合位点,强化了hsa-miR-214-3p作为B7H3调控因子的作用。在乳腺癌中,hsa-miR-214-3p通过下调B7H3抑制肿瘤细胞增殖,增强肿瘤免疫微环境。这些发现为乳腺癌的临床治疗提供了新的潜在靶点。
{"title":"Hsa-miR-214-3p inhibits breast cancer cell growth and improves the tumor immune microenvironment by downregulating B7H3.","authors":"Yan Lu, Kang Wang, Yuanhong Peng, Meng Chen, Lin Zhong, Luji Huang, F U Cheng, Xindan Sheng, Xin Yang, Manzhao Ouyang, George A Calin, Zhiwei He","doi":"10.32604/or.2024.057472","DOIUrl":"10.32604/or.2024.057472","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors play an important role in the treatment of solid tumors, but the currently used immune checkpoint inhibitors targeting programmed cell death-1 (PD-1), programmed cell death ligand-1 (PD-L1), and cytotoxic T-lymphocyte antigen-4 (CTLA-4) show limited clinical efficacy in many breast cancers. B7H3 has been widely reported as an immunosuppressive molecule, but its immunological function in breast cancer patients remains unclear.</p><p><strong>Methods: </strong>We analyzed the expression of B7H3 in breast cancer samples using data from the Cancer Genome Atlas Program (TCGA) and the Gene Expression Omnibus (GEO) databases. MicroRNAs were selected using the TarBase, miRTarBase, and miRBase databases. The regulatory role of the microRNA hsa-miR-214-3p on B7H3 was investigated through dual-luciferase reporter assays, which identified the specific action sites of interaction. The expression levels of B7H3 and hsa-miR-214-3p in human breast cancer tissues and adjacent normal tissues were quantified using Western blotting and quantitative PCR (qPCR). <i>In vitro</i> experiments were performed to observe the effects of modulating the expression of B7H3 or hsa-miR-214-3p on breast cancer cell proliferation and apoptosis. Additionally, the regulatory impact of hsa-miR-214-3p on B7H3 was examined. Enzyme-linked immunosorbent assays (ELISA) and flow cytometry were employed to assess the effects of co-cultured breast cancer cells and normal human peripheral blood mononuclear cells (PBMCs) on immune cells and associated cytokines.</p><p><strong>Results: </strong>In breast cancer tissues, the expression level of B7H3 is inversely correlated with that of hsa-miR-214-3p, as well as with the regulatory effects on breast cancercell behavior. Hsa-miR-214-3p was found to inhibit breast cancer cell growth by downregulating B7H3. Importantly, our research identified, for the first time, two binding sites for hsa-miR-214-3p on the 3' UTR of B7H3, both of which exert similar effects independently. Co-culture experiments revealed that hsa-miR-214-3p obstructs the suppressive function of B7H3 on CD8<sup>+</sup> T cells and natural killer cells.</p><p><strong>Conclusions: </strong>This study confirms the existence of two hsa-miR-214-3p binding sites on the 3' UTR of B7H3, reinforcing the role of hsa-miR-214-3p as a regulatory factor for B7H3. In breast cancer, hsa-miR-214-3p reduces tumor cell proliferation and enhances the tumor immune microenvironment by downregulating B7H3. These findings suggest new potential targets for the clinical treatment of breast cancer.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"103-121"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671619/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903400","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Research advancements in nanoparticles and cell-based drug delivery systems for the targeted killing of cancer cells. 靶向杀伤癌细胞的纳米颗粒和基于细胞的药物传递系统的研究进展。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.056955
Meryem A Abdessalem, Sirin A Adham

Nanotechnology in cancer therapy has significantly advanced treatment precision, effectiveness, and safety, improving patient outcomes and personalized care. Engineered smart nanoparticles and cell-based therapies are designed to target tumor cells, precisely sensing the tumor microenvironment (TME) and sparing normal cells. These nanoparticles enhance drug accumulation in tumors by solubilizing insoluble compounds or preventing their degradation, and they can also overcome therapy resistance and deliver multiple drugs simultaneously. Despite these benefits, challenges remain in patient-specific responses and regulatory approvals for cell-based or nanoparticle therapies. Cell-based drug delivery systems (DDSs) that primarily utilize the immune-recognition principle between ligands and receptors have shown promise in selectively targeting and destroying cancer cells. This review aims to provide a comprehensive overview of various nanoparticle and cell-based drug delivery system types used in cancer research. It covers approved and experimental nanoparticle therapies, including liposomes, micelles, protein-based and polymeric nanoparticles, as well as cell-based DDSs like macrophages, T-lymphocytes, dendritic cells, viruses, bacterial ghosts, minicells, SimCells, and outer membrane vesicles (OMVs). The review also explains the role of TME and its impact on developing smart DDSs in combination therapies and integrating nanoparticles with cell-based systems for targeting cancer cells. By detailing DDSs at different stages of development, from laboratory research to clinical trials and approved treatments, this review provides the latest insights and a collection of valuable citations of the innovative strategies that can be improved for the precise treatment of cancer.

纳米技术在癌症治疗中显著提高了治疗精度、有效性和安全性,改善了患者的治疗效果和个性化护理。工程智能纳米颗粒和基于细胞的疗法被设计用于靶向肿瘤细胞,精确地感知肿瘤微环境(TME)并保留正常细胞。这些纳米颗粒通过溶解不溶性化合物或阻止其降解来增强肿瘤中的药物积累,并且它们还可以克服治疗耐药性并同时递送多种药物。尽管有这些好处,但在细胞或纳米颗粒疗法的患者特异性反应和监管批准方面仍然存在挑战。基于细胞的药物传递系统(dds)主要利用配体和受体之间的免疫识别原理,在选择性靶向和破坏癌细胞方面显示出前景。本综述旨在提供各种纳米颗粒和基于细胞的药物传递系统类型在癌症研究中的全面概述。它涵盖了已批准的和实验性的纳米颗粒疗法,包括脂质体、胶束、蛋白质和聚合物纳米颗粒,以及基于细胞的dds,如巨噬细胞、t淋巴细胞、树突状细胞、病毒、细菌幽灵、微细胞、模拟细胞和外膜囊泡(omv)。这篇综述还解释了TME的作用及其对开发联合治疗中的智能dds和将纳米颗粒与基于细胞的系统结合以靶向癌细胞的影响。通过详细介绍从实验室研究到临床试验和批准治疗的不同发展阶段的dds,本综述提供了最新的见解和有价值的创新策略的集合,可以改进癌症的精确治疗。
{"title":"Research advancements in nanoparticles and cell-based drug delivery systems for the targeted killing of cancer cells.","authors":"Meryem A Abdessalem, Sirin A Adham","doi":"10.32604/or.2024.056955","DOIUrl":"10.32604/or.2024.056955","url":null,"abstract":"<p><p>Nanotechnology in cancer therapy has significantly advanced treatment precision, effectiveness, and safety, improving patient outcomes and personalized care. Engineered smart nanoparticles and cell-based therapies are designed to target tumor cells, precisely sensing the tumor microenvironment (TME) and sparing normal cells. These nanoparticles enhance drug accumulation in tumors by solubilizing insoluble compounds or preventing their degradation, and they can also overcome therapy resistance and deliver multiple drugs simultaneously. Despite these benefits, challenges remain in patient-specific responses and regulatory approvals for cell-based or nanoparticle therapies. Cell-based drug delivery systems (DDSs) that primarily utilize the immune-recognition principle between ligands and receptors have shown promise in selectively targeting and destroying cancer cells. This review aims to provide a comprehensive overview of various nanoparticle and cell-based drug delivery system types used in cancer research. It covers approved and experimental nanoparticle therapies, including liposomes, micelles, protein-based and polymeric nanoparticles, as well as cell-based DDSs like macrophages, T-lymphocytes, dendritic cells, viruses, bacterial ghosts, minicells, SimCells, and outer membrane vesicles (OMVs). The review also explains the role of TME and its impact on developing smart DDSs in combination therapies and integrating nanoparticles with cell-based systems for targeting cancer cells. By detailing DDSs at different stages of development, from laboratory research to clinical trials and approved treatments, this review provides the latest insights and a collection of valuable citations of the innovative strategies that can be improved for the precise treatment of cancer.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"27-44"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671623/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142902853","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Lenalidomide regulates the CCL21/CCR7/ERK1/2 axis to inhibit migration and proliferation in diffuse large B-cell lymphoma. 来那度胺调节CCL21/CCR7/ERK1/2轴抑制弥漫性大b细胞淋巴瘤的迁移和增殖。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.050036
Wen Yang, Bin Tang, Dan Xu, Wenxiu Yang

Background: The prognostic significance of the chemokine receptor CCR7 in diffuse large B-cell lymphoma (DLBCL) has been reported previously. However, the detailed mechanisms of CCR7 in DLBCL, particularly regarding its interaction with lenalidomide treatment, are not fully understood.

Methods: Our study utilized bioinformatics approaches to identify hub genes in SU-DHL-2 cell lines treated with lenalidomide compared to control groups. Immunohistochemical data and clinical information from 122 patients with DLBCL were analyzed to assess the correlation of CCR7 and p-ERK1/2 expression with the prognosis of DLBCL. Furthermore, in vitro and in vivo experiments were conducted to clarify the role of CCR7 in the response of DLBCL to lenalidomide treatment.

Results: Our bioinformatics analysis pinpointed CCR7 as a hub gene in the context of lenalidomide treatment in DLBCL. Notably, 31.14% and 36.0% (44/122) of DLBCL cases showed positive expression for CCR7 and ERK1/2 respectively, establishing them as independent prognostic factors for adverse outcomes in DLBCL via multivariate Cox regression analysis. Additionally, our studies demonstrated that the external application of the protein CCL21 promoted proliferation, migration, invasion, and activation of the ERK1/2 pathway in SU-DHL-2 and OCI-LY3 cell lines with high levels of CCR7 expression. This effect was mitigated by CCR7 silencing through siRNA, application of ERK inhibitors, or lenalidomide treatment. In vivo experiments reinforced the efficacy of lenalidomide, significantly reducing tumor growth rate, tumor mass, serum total LDH levels, and expression of CCR7 and p-ERK1/2 in a SU-DHL-2 xenograft model in nude mice (p < 0.05).

Conclusion: Our study clarifies the potential role of the CCL21/CCR7/ERK1/2 axis in the therapeutic effects of lenalidomide in DLBCL treatment.

背景:趋化因子受体CCR7在弥漫性大b细胞淋巴瘤(DLBCL)中的预后意义已有报道。然而,CCR7在DLBCL中的详细机制,特别是其与来那度胺治疗的相互作用尚不完全清楚。方法:本研究利用生物信息学方法鉴定来那度胺处理的SU-DHL-2细胞系与对照组的中枢基因。我们分析了122例DLBCL患者的免疫组化数据和临床资料,以评估CCR7和p-ERK1/2表达与DLBCL预后的相关性。此外,我们还进行了体外和体内实验,以阐明CCR7在DLBCL对来那度胺治疗反应中的作用。结果:我们的生物信息学分析确定CCR7是来那度胺治疗DLBCL的枢纽基因。值得注意的是,31.14%和36.0%(44/122)的DLBCL病例CCR7和ERK1/2表达阳性,通过多因素Cox回归分析,CCR7和ERK1/2是DLBCL不良结局的独立预后因素。此外,我们的研究表明,在CCR7高表达的SU-DHL-2和OCI-LY3细胞系中,外用CCL21蛋白促进了ERK1/2途径的增殖、迁移、侵袭和激活。通过siRNA沉默CCR7、应用ERK抑制剂或来那度胺治疗可以减轻这种影响。体内实验强化了来那度胺的作用,在SU-DHL-2异种移植裸鼠模型中,来那度胺显著降低肿瘤生长速度、肿瘤体积、血清总LDH水平以及CCR7和p- erk1 /2的表达(p < 0.05)。结论:本研究阐明了CCL21/CCR7/ERK1/2轴在来那度胺治疗DLBCL疗效中的潜在作用。
{"title":"Lenalidomide regulates the CCL21/CCR7/ERK1/2 axis to inhibit migration and proliferation in diffuse large B-cell lymphoma.","authors":"Wen Yang, Bin Tang, Dan Xu, Wenxiu Yang","doi":"10.32604/or.2024.050036","DOIUrl":"10.32604/or.2024.050036","url":null,"abstract":"<p><strong>Background: </strong>The prognostic significance of the chemokine receptor CCR7 in diffuse large B-cell lymphoma (DLBCL) has been reported previously. However, the detailed mechanisms of CCR7 in DLBCL, particularly regarding its interaction with lenalidomide treatment, are not fully understood.</p><p><strong>Methods: </strong>Our study utilized bioinformatics approaches to identify hub genes in SU-DHL-2 cell lines treated with lenalidomide compared to control groups. Immunohistochemical data and clinical information from 122 patients with DLBCL were analyzed to assess the correlation of CCR7 and p-ERK1/2 expression with the prognosis of DLBCL. Furthermore, <i>in vitro</i> and <i>in vivo</i> experiments were conducted to clarify the role of CCR7 in the response of DLBCL to lenalidomide treatment.</p><p><strong>Results: </strong>Our bioinformatics analysis pinpointed CCR7 as a hub gene in the context of lenalidomide treatment in DLBCL. Notably, 31.14% and 36.0% (44/122) of DLBCL cases showed positive expression for CCR7 and ERK1/2 respectively, establishing them as independent prognostic factors for adverse outcomes in DLBCL via multivariate Cox regression analysis. Additionally, our studies demonstrated that the external application of the protein CCL21 promoted proliferation, migration, invasion, and activation of the ERK1/2 pathway in SU-DHL-2 and OCI-LY3 cell lines with high levels of CCR7 expression. This effect was mitigated by CCR7 silencing through siRNA, application of ERK inhibitors, or lenalidomide treatment. <i>In vivo</i> experiments reinforced the efficacy of lenalidomide, significantly reducing tumor growth rate, tumor mass, serum total LDH levels, and expression of CCR7 and p-ERK1/2 in a SU-DHL-2 xenograft model in nude mice (<i>p</i> < 0.05).</p><p><strong>Conclusion: </strong>Our study clarifies the potential role of the CCL21/CCR7/ERK1/2 axis in the therapeutic effects of lenalidomide in DLBCL treatment.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"199-212"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671405/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903405","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
The impact of alpha-fetoprotein (AFP), child-turcotte-pugh (CTP) score and disease staging on the survival of hepatocellular carcinoma (HCC) patients: a retrospective cohort from single oncology center. 甲胎蛋白(AFP)、child- turcote -pugh (CTP)评分和疾病分期对肝细胞癌(HCC)患者生存的影响:来自单个肿瘤中心的回顾性队列研究
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.050903
Nasser Mulla, Yousef Katib, Asim M Almughamsi, Duaa S Alkhayat, Mohamed Mosaad, Samir T Alfotih, Rawan Alaofi

Background: Hepatocellular carcinoma (HCC) is the most common cause of cancer-related death in Saudi Arabia. Our study aimed to investigate the patterns of HCC and the effect of TNM staging, Alfa-fetoprotein (AFP), and Child-Turcotte Pugh (CTP) on patients' overall survival (OS).

Methods: A retrospective analysis was conducted on 43 HCC patients at a single oncology center in Saudi Arabia from 2015 to 2020. All patients had to fulfill one of the following criteria: (a) a liver lesion reported as definitive HCC on dynamic imaging and/or (b) a biopsy-confirmed diagnosis.

Results: The mean patient age of all HCC cases was 66.8 with a male-to-female ratio of 3.3:1. All patients were stratified into two groups: viral HCC (n = 22, 51%) and non-viral HCC (n = 21, 49%). Among viral-HCC patients, 55% were due to HBV and 45% due to HCV. Cirrhosis was diagnosed in 79% of cases. Age and sex did not significantly statistically differ in OS among viral and non-viral HCC patients (p-value > 0.05). About 65% of patients had tumor size >5 cm during the diagnosis, with a significant statistical difference in OS (p-value = 0.027). AFP was >400 ng/ml in 45% of the patients. There was a statistically significant difference in the OS in terms of AFP levels (p-value = 0.021). A statistically significant difference was also observed between the CTP score and OS (p-value = 0.02). CTP class B had the longest survival. BSC was the most common treatment provided to HCC patients followed by sorafenib therapy. There was a significant statistical difference in OS among viral and non-viral HCC patients (p-value = 0.008).

Conclusions: The most common predictors for OS were the underlying cause of HCC, AFP, and tumor size. Being having non-viral etiology, a tumor size >5 cm, an AFP > 400 ng/mL, and a CTP score class C were all negatively associated with OS.

背景:肝细胞癌(HCC)是沙特阿拉伯癌症相关死亡的最常见原因。本研究旨在探讨HCC的类型以及TNM分期、甲胎蛋白(AFP)和Child-Turcotte Pugh (CTP)对患者总生存期(OS)的影响。方法:对沙特阿拉伯某肿瘤中心2015 - 2020年收治的43例HCC患者进行回顾性分析。所有患者必须满足以下标准之一:(a)动态影像学报告的肝脏病变为明确的HCC和/或(b)活检证实的诊断。结果:所有HCC患者的平均年龄为66.8岁,男女比例为3.3:1。所有患者被分为两组:病毒性HCC (n = 22, 51%)和非病毒性HCC (n = 21, 49%)。在病毒性hcc患者中,55%是HBV所致,45%是HCV所致。79%的病例被诊断为肝硬化。病毒性和非病毒性HCC患者OS的年龄、性别差异无统计学意义(p值0.05)。约65%的患者诊断时肿瘤大小为bbb5 cm, OS差异有统计学意义(p值= 0.027)。45%的患者AFP为100 ~ 400 ng/ml。两组间甲胎蛋白水平差异有统计学意义(p值= 0.021)。CTP评分与OS的差异也有统计学意义(p值= 0.02)。CTP B类患者存活时间最长。BSC是HCC患者最常见的治疗方法,其次是索拉非尼治疗。病毒性和非病毒性HCC患者的OS差异有统计学意义(p值= 0.008)。结论:最常见的OS预测因子是HCC的潜在原因、AFP和肿瘤大小。非病毒性病因、肿瘤大小bbbb5 cm、AFP b> 400 ng/mL、CTP评分C级均与OS呈负相关。
{"title":"The impact of alpha-fetoprotein (AFP), child-turcotte-pugh (CTP) score and disease staging on the survival of hepatocellular carcinoma (HCC) patients: a retrospective cohort from single oncology center.","authors":"Nasser Mulla, Yousef Katib, Asim M Almughamsi, Duaa S Alkhayat, Mohamed Mosaad, Samir T Alfotih, Rawan Alaofi","doi":"10.32604/or.2024.050903","DOIUrl":"10.32604/or.2024.050903","url":null,"abstract":"<p><strong>Background: </strong>Hepatocellular carcinoma (HCC) is the most common cause of cancer-related death in Saudi Arabia. Our study aimed to investigate the patterns of HCC and the effect of TNM staging, Alfa-fetoprotein (AFP), and Child-Turcotte Pugh (CTP) on patients' overall survival (OS).</p><p><strong>Methods: </strong>A retrospective analysis was conducted on 43 HCC patients at a single oncology center in Saudi Arabia from 2015 to 2020. All patients had to fulfill one of the following criteria: (a) a liver lesion reported as definitive HCC on dynamic imaging and/or (b) a biopsy-confirmed diagnosis.</p><p><strong>Results: </strong>The mean patient age of all HCC cases was 66.8 with a male-to-female ratio of 3.3:1. All patients were stratified into two groups: viral HCC (n = 22, 51%) and non-viral HCC (n = 21, 49%). Among viral-HCC patients, 55% were due to HBV and 45% due to HCV. Cirrhosis was diagnosed in 79% of cases. Age and sex did not significantly statistically differ in OS among viral and non-viral HCC patients (<i>p</i>-value > 0.05). About 65% of patients had tumor size >5 cm during the diagnosis, with a significant statistical difference in OS (<i>p</i>-value = 0.027). AFP was >400 ng/ml in 45% of the patients. There was a statistically significant difference in the OS in terms of AFP levels (<i>p</i>-value = 0.021). A statistically significant difference was also observed between the CTP score and OS (<i>p</i>-value = 0.02). CTP class B had the longest survival. BSC was the most common treatment provided to HCC patients followed by sorafenib therapy. There was a significant statistical difference in OS among viral and non-viral HCC patients (<i>p</i>-value = 0.008).</p><p><strong>Conclusions: </strong>The most common predictors for OS were the underlying cause of HCC, AFP, and tumor size. Being having non-viral etiology, a tumor size >5 cm, an AFP > 400 ng/mL, and a CTP score class C were all negatively associated with OS.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"149-160"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671413/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903141","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Three-dimensional models: from cell culture to Patient-Derived Organoid and its application to future liposarcoma research. 三维模型:从细胞培养到患者来源的类器官及其在未来脂肪肉瘤研究中的应用。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.053635
Sayumi Tahara, Sydney Rentsch, Fernanda Costas Casal DE Faria, Patricia Sarchet, Roma Karna, Federica Calore, Raphael E Pollock

Liposarcoma is one of the most common soft tissue sarcomas, however, its occurrence rate is still rare compared to other cancers. Due to its rarity, in vitro experiments are an essential approach to elucidate liposarcoma pathobiology. Conventional cell culture-based research (2D cell culture) is still playing a pivotal role, while several shortcomings have been recently under discussion. In vivo, mouse models are usually adopted for pre-clinical analyses with expectations to overcome the issues of 2D cell culture. However, they do not fully recapitulate human dedifferentiated liposarcoma (DDLPS) characteristics. Therefore, three-dimensional (3D) culture systems have been the recent research focus in the cell biology field with the expectation to overcome at the same time the disadvantages of 2D cell culture and in vivo animal models and fill in the gap between them. Given the liposarcoma rarity, we believe that 3D cell culture techniques, including 3D cell cultures/co-cultures, and Patient-Derived tumor Organoids (PDOs), represent a promising approach to facilitate liposarcoma investigation and elucidate its molecular mechanisms and effective therapy development. In this review, we first provide a general overview of 3D cell cultures compared to 2D cell cultures. We then focus on one of the recent 3D cell culture applications, Patient-Derived Organoids (PDOs), summarizing and discussing several PDO methodologies. Finally, we discuss the current and future applications of PDOs to sarcoma, particularly in the field of liposarcoma.

脂肪肉瘤是最常见的软组织肉瘤之一,但与其他肿瘤相比,其发病率仍属罕见。由于其罕见性,体外实验是阐明脂肪肉瘤病理生物学的重要途径。传统的以细胞培养为基础的研究(二维细胞培养)仍然发挥着关键作用,然而最近人们正在讨论一些缺点。在体内,通常采用小鼠模型进行临床前分析,以期克服二维细胞培养的问题。然而,它们并不能完全概括人去分化脂肪肉瘤(DDLPS)的特征。因此,三维(3D)培养系统一直是近年来细胞生物学领域的研究热点,希望能同时克服二维细胞培养和活体动物模型的不足,填补两者之间的空白。鉴于脂肪肉瘤的罕见性,我们认为3D细胞培养技术,包括3D细胞培养/共培养和患者源性肿瘤类器官(PDOs),代表了促进脂肪肉瘤研究和阐明其分子机制和有效治疗开发的有前途的方法。在这篇综述中,我们首先提供了3D细胞培养与2D细胞培养的一般概述。然后,我们专注于最近的3D细胞培养应用之一,患者衍生类器官(PDO),总结和讨论了几种PDO方法。最后,我们讨论了PDOs在肉瘤中的应用现状和未来,特别是在脂肪肉瘤领域。
{"title":"Three-dimensional models: from cell culture to Patient-Derived Organoid and its application to future liposarcoma research.","authors":"Sayumi Tahara, Sydney Rentsch, Fernanda Costas Casal DE Faria, Patricia Sarchet, Roma Karna, Federica Calore, Raphael E Pollock","doi":"10.32604/or.2024.053635","DOIUrl":"10.32604/or.2024.053635","url":null,"abstract":"<p><p>Liposarcoma is one of the most common soft tissue sarcomas, however, its occurrence rate is still rare compared to other cancers. Due to its rarity, <i>in vitro</i> experiments are an essential approach to elucidate liposarcoma pathobiology. Conventional cell culture-based research (2D cell culture) is still playing a pivotal role, while several shortcomings have been recently under discussion. <i>In vivo</i>, mouse models are usually adopted for pre-clinical analyses with expectations to overcome the issues of 2D cell culture. However, they do not fully recapitulate human dedifferentiated liposarcoma (DDLPS) characteristics. Therefore, three-dimensional (3D) culture systems have been the recent research focus in the cell biology field with the expectation to overcome at the same time the disadvantages of 2D cell culture and <i>in vivo</i> animal models and fill in the gap between them. Given the liposarcoma rarity, we believe that 3D cell culture techniques, including 3D cell cultures/co-cultures, and Patient-Derived tumor Organoids (PDOs), represent a promising approach to facilitate liposarcoma investigation and elucidate its molecular mechanisms and effective therapy development. In this review, we first provide a general overview of 3D cell cultures compared to 2D cell cultures. We then focus on one of the recent 3D cell culture applications, Patient-Derived Organoids (PDOs), summarizing and discussing several PDO methodologies. Finally, we discuss the current and future applications of PDOs to sarcoma, particularly in the field of liposarcoma.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"1-13"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671414/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903156","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Engendered nanoparticles for treatment of brain tumors. 人造纳米颗粒治疗脑肿瘤。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.053069
Soroush Soleymani, Mohammad Doroudian, Mahdieh Soezi, Ali Beladi, Kiarash Asgari, Aso Mobarakshahi, Aryana Aghaeipour, Ronan Macloughlin

Brain metastasis and primary glioblastoma multiforme represent the most common and lethal malignant brain tumors. Its median survival time is typically less than a year after diagnosis. One of the major challenges in treating these cancers is the efficiency of the transport of drugs to the central nervous system. The blood-brain barrier is cooperating with advanced stages of malignancy. The blood-brain barrier poses a significant challenge to delivering systemic medications to brain tumors. Nanodrug delivery systems have emerged as promising tools for effectively crossing this barrier. Additionally, the development of smart nanoparticles brings new hope for cancer diagnosis and treatment. These nanoparticles improve drug delivery efficiency, allowing for the creation of targeted and stimuli-responsive delivery methods. This review highlights recent advancements in nanoparticle and smart nanoparticle technologies for brain cancer treatment, exploring the range of nanoparticles under development, their applications, targeting strategies, and the latest progress in enhancing transport across the blood-brain barrier. It also addresses the ongoing challenges and potential benefits of these innovative approaches.

脑转移瘤和原发性多形性胶质母细胞瘤是最常见和最致命的恶性脑肿瘤。它的中位生存时间通常在诊断后不到一年。治疗这些癌症的主要挑战之一是药物转运到中枢神经系统的效率。血脑屏障与恶性肿瘤的晚期相配合。血脑屏障对向脑肿瘤输送全身药物构成了重大挑战。纳米药物输送系统已经成为有效跨越这一屏障的有前途的工具。此外,智能纳米粒子的发展为癌症的诊断和治疗带来了新的希望。这些纳米颗粒提高了药物递送效率,允许创建靶向和刺激反应的递送方法。本文重点介绍了纳米颗粒和智能纳米颗粒技术在脑癌治疗中的最新进展,探讨了纳米颗粒的开发范围、应用、靶向策略以及增强血脑屏障运输的最新进展。它还讨论了这些创新方法的持续挑战和潜在好处。
{"title":"Engendered nanoparticles for treatment of brain tumors.","authors":"Soroush Soleymani, Mohammad Doroudian, Mahdieh Soezi, Ali Beladi, Kiarash Asgari, Aso Mobarakshahi, Aryana Aghaeipour, Ronan Macloughlin","doi":"10.32604/or.2024.053069","DOIUrl":"10.32604/or.2024.053069","url":null,"abstract":"<p><p>Brain metastasis and primary glioblastoma multiforme represent the most common and lethal malignant brain tumors. Its median survival time is typically less than a year after diagnosis. One of the major challenges in treating these cancers is the efficiency of the transport of drugs to the central nervous system. The blood-brain barrier is cooperating with advanced stages of malignancy. The blood-brain barrier poses a significant challenge to delivering systemic medications to brain tumors. Nanodrug delivery systems have emerged as promising tools for effectively crossing this barrier. Additionally, the development of smart nanoparticles brings new hope for cancer diagnosis and treatment. These nanoparticles improve drug delivery efficiency, allowing for the creation of targeted and stimuli-responsive delivery methods. This review highlights recent advancements in nanoparticle and smart nanoparticle technologies for brain cancer treatment, exploring the range of nanoparticles under development, their applications, targeting strategies, and the latest progress in enhancing transport across the blood-brain barrier. It also addresses the ongoing challenges and potential benefits of these innovative approaches.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"15-26"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671406/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903394","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Long noncoding RNA LINC01106 promotes lung adenocarcinoma progression via upregulation of autophagy. 长链非编码RNA LINC01106通过上调自噬促进肺腺癌进展。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.047626
Gengyun Sun, Yiping Zheng, Jianfeng Cai, Jie Gao, Lie Dong, Xiangbin Zhang, Yinghui Huang

Background: Long noncoding RNA, LINC01106 exhibits high expression in lung adenocarcinoma (LUAD) tumor tissues, but its functional role and regulatory mechanism in LUAD cells remain unclear.

Methods: LINC01106 expression was analyzed in LUAD tissues and its functional impact on LUAD cells was assessed. LUAD cells were silenced with sh-LINC01106 and injected into nude mice to investigate tumor growth. The downstream transcription factors and molecular mechanism were determined using the Human transcription factor database (TFDB) database and Gene Expression Profiling Interactive Analysis (GEPIA) database. Additionally, the impact of linc01106 on autophagy was analyzed by determining the expression of autophagy-related genes (ATGs) in LUAD cells.

Results: Our results showed that LINC01106 exhibited upregulation in both LUAD tissues and cell lines. The silencing of LINC01106 demonstrated a suppressive effect on tumorigenesis in a xenograft mouse model of LUAD. Additionally, LINC01106 was found to recruit TATA-binding protein-associated factor 15 (TAF15), an RNA-binding protein, thereby enhancing the mRNA stability of TEA domain transcription factor 4 (TEAD4). In turn, TEAD4 served as a transcription factor that bound to the LINC01106 promoter and regulated its expression. Further assays indicated that LINC01106 promoted autophagy in LUAD cells by upregulating the expression of autophagy-related genes (ATGs). The silencing of LINC01106 in LUAD cells inhibited autophagy, and cell proliferation, and promoted apoptosis, which all were effectively reversed by ATG5 overexpression.

Conclusions: Overall, LINC01106, transcriptionally activated by TEAD4, interacts with TAF15 to promote the stability of TEAD4 and upregulates the expression of ATGs, promoting malignancy of LUAD cells.

背景:长链非编码RNA LINC01106在肺腺癌(LUAD)肿瘤组织中高表达,但其在LUAD细胞中的功能作用和调控机制尚不清楚。方法:分析LINC01106在LUAD组织中的表达,评估其对LUAD细胞功能的影响。用sh-LINC01106沉默LUAD细胞,并注射到裸鼠体内,观察肿瘤的生长情况。利用Human transcription factor database (TFDB)数据库和Gene Expression Profiling Interactive Analysis (GEPIA)数据库确定下游转录因子及其分子机制。此外,通过检测LUAD细胞中自噬相关基因(autophagy-related genes, ATGs)的表达,分析linc01106对自噬的影响。结果:我们的研究结果表明,LINC01106在LUAD组织和细胞系中均表现出上调。在异种移植小鼠LUAD模型中,沉默LINC01106显示出抑制肿瘤发生的作用。此外,我们发现LINC01106募集了一种rna结合蛋白tata binding protein associated factor 15 (TAF15),从而增强了TEA domain transcription factor 4 (TEAD4) mRNA的稳定性。反过来,TEAD4作为转录因子结合到LINC01106启动子并调节其表达。进一步的实验表明,LINC01106通过上调自噬相关基因(autophagy-related genes, ATGs)的表达,促进LUAD细胞的自噬。LINC01106在LUAD细胞中的沉默抑制了细胞自噬,抑制了细胞增殖,促进了细胞凋亡,这些都被ATG5过表达有效逆转。结论:总的来说,被TEAD4转录激活的LINC01106与TAF15相互作用,促进TEAD4的稳定性,上调ATGs的表达,促进LUAD细胞的恶性化。
{"title":"Long noncoding RNA LINC01106 promotes lung adenocarcinoma progression via upregulation of autophagy.","authors":"Gengyun Sun, Yiping Zheng, Jianfeng Cai, Jie Gao, Lie Dong, Xiangbin Zhang, Yinghui Huang","doi":"10.32604/or.2024.047626","DOIUrl":"10.32604/or.2024.047626","url":null,"abstract":"<p><strong>Background: </strong>Long noncoding RNA, LINC01106 exhibits high expression in lung adenocarcinoma (LUAD) tumor tissues, but its functional role and regulatory mechanism in LUAD cells remain unclear.</p><p><strong>Methods: </strong>LINC01106 expression was analyzed in LUAD tissues and its functional impact on LUAD cells was assessed. LUAD cells were silenced with sh-LINC01106 and injected into nude mice to investigate tumor growth. The downstream transcription factors and molecular mechanism were determined using the Human transcription factor database (TFDB) database and Gene Expression Profiling Interactive Analysis (GEPIA) database. Additionally, the impact of linc01106 on autophagy was analyzed by determining the expression of autophagy-related genes (ATGs) in LUAD cells.</p><p><strong>Results: </strong>Our results showed that LINC01106 exhibited upregulation in both LUAD tissues and cell lines. The silencing of LINC01106 demonstrated a suppressive effect on tumorigenesis in a xenograft mouse model of LUAD. Additionally, LINC01106 was found to recruit TATA-binding protein-associated factor 15 (TAF15), an RNA-binding protein, thereby enhancing the mRNA stability of TEA domain transcription factor 4 (TEAD4). In turn, TEAD4 served as a transcription factor that bound to the LINC01106 promoter and regulated its expression. Further assays indicated that LINC01106 promoted autophagy in LUAD cells by upregulating the expression of autophagy-related genes (ATGs). The silencing of LINC01106 in LUAD cells inhibited autophagy, and cell proliferation, and promoted apoptosis, which all were effectively reversed by ATG5 overexpression.</p><p><strong>Conclusions: </strong>Overall, LINC01106, transcriptionally activated by TEAD4, interacts with TAF15 to promote the stability of TEAD4 and upregulates the expression of ATGs, promoting malignancy of LUAD cells.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"171-184"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671404/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903411","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
TMED3 promotes prostate cancer via FOXO1a and FOXO3a phosphorylation. TMED3通过FOXO1a和FOXO3a磷酸化促进前列腺癌。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.048054
Xiuwang Wei, Jianbo Liang, Huanwen Huang, Daming Yang, Xinxin Wang, Xiujia Wang, Changsheng Chen, Kaiqiang Li, Taisen Pang, Bin Hu, Fengning Wu

Background: Transmembrane emp24 trafficking protein 3 (TMED3) is associated with the development of several tumors; however, whether TMED3 regulates the progression of prostate cancer remains unclear.

Materials and methods: Short hairpin RNA was performed to repress TMED3 in prostate cancer cells (DU145 cells) and in a prostate cancer mice model to determine its function in prostate cancer in vitro and in vivo.

Results: In the present study, we found that TMED3 was highly expressed in prostate cancer cells. In vitro, shTMED3 treatment suppressed the proliferation, invasion, and migration and promoted the apoptosis of DU145 cells. Additionally, the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed a strong correlation between TMED3 and forkhead box O transcription factor (FOXO) pathway. Furthermore, TMED3 inhibition efficiently decreased FOXO1a and FOXO3a phosphorylation. In vivo, TMED3 downregulation suppressed the apoptosis, growth, and metastasis of prostate cancer cells via FOXO1a and FOXO3a.

Conclusion: The present findings show that TMED3 participates in the regulation of prostate cancer progression via FOXO1a and FOXO3a phosphorylation, thereby revealing a novel mechanism underlying prostate cancer development and suggesting that TMED3 inhibition may serve as a novel strategy for prostate cancer treatment.

背景:跨膜emp24转运蛋白3 (TMED3)与几种肿瘤的发展有关;然而,TMED3是否调节前列腺癌的进展尚不清楚。材料和方法:利用短发夹RNA在前列腺癌细胞(DU145细胞)和前列腺癌小鼠模型中抑制TMED3,研究其在前列腺癌体内外的功能。结果:在本研究中,我们发现TMED3在前列腺癌细胞中高表达。在体外,shTMED3处理可抑制DU145细胞的增殖、侵袭和迁移,促进DU145细胞的凋亡。此外,京都基因和基因组百科全书通路富集分析显示TMED3与叉头盒O转录因子(FOXO)通路有很强的相关性。此外,TMED3抑制有效降低了FOXO1a和FOXO3a的磷酸化。在体内,TMED3下调通过FOXO1a和FOXO3a抑制前列腺癌细胞的凋亡、生长和转移。结论:本研究结果表明,TMED3通过FOXO1a和FOXO3a磷酸化参与前列腺癌进展的调控,从而揭示了前列腺癌发展的新机制,提示抑制TMED3可能作为前列腺癌治疗的新策略。
{"title":"TMED3 promotes prostate cancer via FOXO1a and FOXO3a phosphorylation.","authors":"Xiuwang Wei, Jianbo Liang, Huanwen Huang, Daming Yang, Xinxin Wang, Xiujia Wang, Changsheng Chen, Kaiqiang Li, Taisen Pang, Bin Hu, Fengning Wu","doi":"10.32604/or.2024.048054","DOIUrl":"10.32604/or.2024.048054","url":null,"abstract":"<p><strong>Background: </strong>Transmembrane emp24 trafficking protein 3 (TMED3) is associated with the development of several tumors; however, whether TMED3 regulates the progression of prostate cancer remains unclear.</p><p><strong>Materials and methods: </strong>Short hairpin RNA was performed to repress TMED3 in prostate cancer cells (DU145 cells) and in a prostate cancer mice model to determine its function in prostate cancer <i>in vitro</i> and <i>in vivo</i>.</p><p><strong>Results: </strong>In the present study, we found that TMED3 was highly expressed in prostate cancer cells. <i>In vitro</i>, shTMED3 treatment suppressed the proliferation, invasion, and migration and promoted the apoptosis of DU145 cells. Additionally, the Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed a strong correlation between TMED3 and forkhead box O transcription factor (FOXO) pathway. Furthermore, TMED3 inhibition efficiently decreased FOXO1a and FOXO3a phosphorylation. <i>In vivo</i>, TMED3 downregulation suppressed the apoptosis, growth, and metastasis of prostate cancer cells via FOXO1a and FOXO3a.</p><p><strong>Conclusion: </strong>The present findings show that TMED3 participates in the regulation of prostate cancer progression via FOXO1a and FOXO3a phosphorylation, thereby revealing a novel mechanism underlying prostate cancer development and suggesting that TMED3 inhibition may serve as a novel strategy for prostate cancer treatment.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"161-169"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671412/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903270","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Ubiquitin-specific protease 1 facilitates tumor immune escape from natural killer cells and predicts the prognosis in small cell lung cancer. 泛素特异性蛋白酶1促进肿瘤对自然杀伤细胞的免疫逃逸并预测小细胞肺癌的预后。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.046895
Shiqin Jiang, Yichun Tang, Feng Ma, Yuchun Niu, Lei Sun

Objective: Small cell lung cancer (SCLC) is commonly recognized as the most fatal lung cancer type. Despite substantial advances in immune checkpoint blockade therapies for treating solid cancers, their benefits are limited to a minority of patients with SCLC. In the present study, novel indicators for predicting the outcomes and molecular targets for SCLC treatment were elucidated.

Methods: We conducted bioinformatics analysis to identify the key genes associated with tumor-infiltrating lymphocytes in SCLC. The functional role of the key gene identified in SCLC was determined both in vitro and in vivo.

Results: A significant correlation was observed between patient survival and CD56dim natural killer (NK) cell proportion. Furthermore, we noted that the hub gene ubiquitin-specific protease 1 (USP1) is closely correlated with both CD56dim NK cells and overall survival in SCLC. Bioinformatics analysis revealed that USP1 is upregulated in SCLC. In addition, gene set enrichment analysis revealed that USP1 overexpression hinders NK cell-mediated immune responses. By co-cultivating NK-92 cells with SCLC cells, we demonstrated that NK cell cytotoxicity against SCLC could be improved either via USP1 knock-down or pharmacological inhibition. Furthermore, using a nude-mice xenograft tumor model, we noted that USP1 inhibition effectively suppressed tumor proliferation and increased the expression of NK cell-associated markers.

Conclusions: Our study findings highlight the importance of NK cells in regulating SCLC. USP1 overexpression can inhibit NK cell-mediated immunity; therefore, USP1 may serve not only as a prognostic biomarker but also as a potential molecular target of SCLC therapy.

目的:小细胞肺癌(SCLC)是公认的最致命的肺癌类型。尽管免疫检查点阻断疗法在治疗实体癌方面取得了实质性进展,但其益处仅限于少数SCLC患者。本研究阐明了预测SCLC治疗结果和分子靶点的新指标。方法:通过生物信息学分析,鉴定SCLC中与肿瘤浸润淋巴细胞相关的关键基因。在体外和体内均确定了SCLC中鉴定的关键基因的功能作用。结果:患者的生存与CD56dim自然杀伤细胞(NK)比例有显著相关性。此外,我们注意到枢纽基因泛素特异性蛋白酶1 (USP1)与SCLC的CD56dim NK细胞和总生存率密切相关。生物信息学分析显示,USP1在SCLC中表达上调。此外,基因集富集分析显示,USP1过表达阻碍NK细胞介导的免疫反应。通过将NK-92细胞与SCLC细胞共培养,我们证明NK细胞对SCLC的细胞毒性可以通过敲除USP1或药物抑制来提高。此外,使用裸鼠异种移植肿瘤模型,我们注意到USP1抑制有效地抑制肿瘤增殖并增加NK细胞相关标记物的表达。结论:我们的研究结果强调了NK细胞在调节SCLC中的重要性。USP1过表达可抑制NK细胞介导的免疫;因此,USP1不仅可以作为预后生物标志物,还可以作为SCLC治疗的潜在分子靶点。
{"title":"Ubiquitin-specific protease 1 facilitates tumor immune escape from natural killer cells and predicts the prognosis in small cell lung cancer.","authors":"Shiqin Jiang, Yichun Tang, Feng Ma, Yuchun Niu, Lei Sun","doi":"10.32604/or.2024.046895","DOIUrl":"10.32604/or.2024.046895","url":null,"abstract":"<p><strong>Objective: </strong>Small cell lung cancer (SCLC) is commonly recognized as the most fatal lung cancer type. Despite substantial advances in immune checkpoint blockade therapies for treating solid cancers, their benefits are limited to a minority of patients with SCLC. In the present study, novel indicators for predicting the outcomes and molecular targets for SCLC treatment were elucidated.</p><p><strong>Methods: </strong>We conducted bioinformatics analysis to identify the key genes associated with tumor-infiltrating lymphocytes in SCLC. The functional role of the key gene identified in SCLC was determined both <i>in vitro</i> and <i>in vivo</i>.</p><p><strong>Results: </strong>A significant correlation was observed between patient survival and CD56dim natural killer (NK) cell proportion. Furthermore, we noted that the hub gene ubiquitin-specific protease 1 (USP1) is closely correlated with both CD56dim NK cells and overall survival in SCLC. Bioinformatics analysis revealed that USP1 is upregulated in SCLC. In addition, gene set enrichment analysis revealed that USP1 overexpression hinders NK cell-mediated immune responses. By co-cultivating NK-92 cells with SCLC cells, we demonstrated that NK cell cytotoxicity against SCLC could be improved either via USP1 knock-down or pharmacological inhibition. Furthermore, using a nude-mice xenograft tumor model, we noted that USP1 inhibition effectively suppressed tumor proliferation and increased the expression of NK cell-associated markers.</p><p><strong>Conclusions: </strong>Our study findings highlight the importance of NK cells in regulating SCLC. USP1 overexpression can inhibit NK cell-mediated immunity; therefore, USP1 may serve not only as a prognostic biomarker but also as a potential molecular target of SCLC therapy.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"213-224"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671408/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903271","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
CAF-derived exosome-miR-3124-5p promotes malignant biological processes in NSCLC via the TOLLIP/TLR4-MyD88-NF-κB pathway. cafa衍生的外泌体- mir -3124-5p通过TOLLIP/TLR4-MyD88-NF-κB途径促进NSCLC的恶性生物学过程。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.054141
Tao Sun, Qinghua Song, Hua Liu

Background: Lung cancer is a life-threatening disease that occurs worldwide, but is especially common in China. The crucial role of the tumour microenvironment (TME) in non-small cell lung cancer (NSCLC) has attracted recent attention. Cancer-associated fibroblasts (CAFs) are the main factors that contribute to the TME function, and CAF exosomes are closely linked to NSCLC.

Methods: The expression levels of miR-3124-5p and Toll-interacting protein (TOLLIP) were analysed by bioinformatics prediction combined with RT-qPCR/Western Blot detection. Fibroblasts were isolated and identified from clinical NSCLC tissues. Transmission electron microscopy and Western Blot were used to identify exosomes from these cells. Changes in proliferation (CCK-8 and clone formation), migration (wound healing), and invasion (transwell) of NSCLC cells were measured. The Luciferase reporter test was applied to clarify the binding of miR-3124-5p to TOLLIP. The TOLLIP/TLR4/MyD88/NF-κB pathway proteins were determined using Western blot analysis.

Results: MiR-3124-5p is overexpressed in clinical tissues and cells of NSCLC. MiR-3124-5p was dramatically enriched in CAF-derived exosomes. Cellular experiments revealed that CAFs delivered miR-3124-5p into NSCLC cells via exosomes, stimulating cancer cell progression. MiR-3124-5p acted as a sponge to negatively regulate TOLLIP expression, which activated the TLR4/MyD88/NF-κB axis to promote the occurrence and development of NSCLC. Functional salvage tests were performed to determine whether CAF-exosome-derived miR-3124-5p plays a pro-cancer role in NSCLC by affecting the TOLLIP signalling pathway.

Conclusions: These results provide an interesting direction for the diagnosis and therapy of NSCLC.

背景:肺癌是一种全球性的危及生命的疾病,但在中国尤为常见。肿瘤微环境(TME)在非小细胞肺癌(NSCLC)中的重要作用近年来引起了人们的关注。癌症相关成纤维细胞(CAFs)是TME功能的主要因素,CAF外泌体与NSCLC密切相关。方法:采用生物信息学预测结合RT-qPCR/Western Blot检测,分析miR-3124-5p和toll相互作用蛋白(TOLLIP)的表达水平。从临床非小细胞肺癌组织中分离鉴定成纤维细胞。利用透射电镜和Western Blot技术鉴定这些细胞的外泌体。测量非小细胞肺癌细胞增殖(CCK-8和克隆形成)、迁移(伤口愈合)和侵袭(transwell)的变化。应用荧光素酶报告基因试验阐明miR-3124-5p与TOLLIP的结合。Western blot检测TOLLIP/TLR4/MyD88/NF-κB通路蛋白表达。结果:MiR-3124-5p在非小细胞肺癌临床组织和细胞中过表达。MiR-3124-5p在caf衍生的外泌体中显著富集。细胞实验显示,CAFs通过外泌体将miR-3124-5p传递到NSCLC细胞中,刺激癌细胞进展。MiR-3124-5p作为海绵负向调控TOLLIP表达,激活TLR4/MyD88/NF-κB轴,促进NSCLC的发生发展。进行功能挽救试验,以确定ca -外泌体来源的miR-3124-5p是否通过影响TOLLIP信号通路在NSCLC中发挥促癌作用。结论:这些结果为非小细胞肺癌的诊断和治疗提供了一个有意义的方向。
{"title":"CAF-derived exosome-miR-3124-5p promotes malignant biological processes in NSCLC via the TOLLIP/TLR4-MyD88-NF-κB pathway.","authors":"Tao Sun, Qinghua Song, Hua Liu","doi":"10.32604/or.2024.054141","DOIUrl":"10.32604/or.2024.054141","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer is a life-threatening disease that occurs worldwide, but is especially common in China. The crucial role of the tumour microenvironment (TME) in non-small cell lung cancer (NSCLC) has attracted recent attention. Cancer-associated fibroblasts (CAFs) are the main factors that contribute to the TME function, and CAF exosomes are closely linked to NSCLC.</p><p><strong>Methods: </strong>The expression levels of miR-3124-5p and Toll-interacting protein (TOLLIP) were analysed by bioinformatics prediction combined with RT-qPCR/Western Blot detection. Fibroblasts were isolated and identified from clinical NSCLC tissues. Transmission electron microscopy and Western Blot were used to identify exosomes from these cells. Changes in proliferation (CCK-8 and clone formation), migration (wound healing), and invasion (transwell) of NSCLC cells were measured. The Luciferase reporter test was applied to clarify the binding of miR-3124-5p to TOLLIP. The TOLLIP/TLR4/MyD88/NF-κB pathway proteins were determined using Western blot analysis.</p><p><strong>Results: </strong>MiR-3124-5p is overexpressed in clinical tissues and cells of NSCLC. MiR-3124-5p was dramatically enriched in CAF-derived exosomes. Cellular experiments revealed that CAFs delivered miR-3124-5p into NSCLC cells via exosomes, stimulating cancer cell progression. MiR-3124-5p acted as a sponge to negatively regulate TOLLIP expression, which activated the TLR4/MyD88/NF-κB axis to promote the occurrence and development of NSCLC. Functional salvage tests were performed to determine whether CAF-exosome-derived miR-3124-5p plays a pro-cancer role in NSCLC by affecting the TOLLIP signalling pathway.</p><p><strong>Conclusions: </strong>These results provide an interesting direction for the diagnosis and therapy of NSCLC.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"133-148"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671617/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
期刊
Oncology Research
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