首页 > 最新文献

Cellular Oncology最新文献

英文 中文
Roles for macrophage-polarizing interleukins in cancer immunity and immunotherapy 巨噬细胞极化白介素在癌症免疫和免疫治疗中的作用
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2022-05-19 DOI: 10.1007/s13402-022-00667-8
K. Mortezaee, Jamal Majidpoor
{"title":"Roles for macrophage-polarizing interleukins in cancer immunity and immunotherapy","authors":"K. Mortezaee, Jamal Majidpoor","doi":"10.1007/s13402-022-00667-8","DOIUrl":"https://doi.org/10.1007/s13402-022-00667-8","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2022-05-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45135798","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: 23
Expression of NOTCH1, NOTCH4, HLA-DMA and HLA-DRA is synergistically associated with T cell exclusion, immune checkpoint blockade efficacy and recurrence risk in ER-negative breast cancer 在er阴性乳腺癌中,NOTCH1、NOTCH4、HLA-DMA和HLA-DRA的表达与T细胞排斥、免疫检查点阻断疗效和复发风险协同相关
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2022-05-11 DOI: 10.1007/s13402-022-00677-6
Dingxie Liu, P. Hofman
{"title":"Expression of NOTCH1, NOTCH4, HLA-DMA and HLA-DRA is synergistically associated with T cell exclusion, immune checkpoint blockade efficacy and recurrence risk in ER-negative breast cancer","authors":"Dingxie Liu, P. Hofman","doi":"10.1007/s13402-022-00677-6","DOIUrl":"https://doi.org/10.1007/s13402-022-00677-6","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2022-05-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45890012","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: 3
LINC02154 promotes the proliferation and metastasis of hepatocellular carcinoma by enhancing SPC24 promoter activity and activating the PI3K-AKT signaling pathway LINC02154通过增强SPC24启动子活性和激活PI3K-AKT信号通路促进肝癌的增殖和转移
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2022-05-11 DOI: 10.1007/s13402-022-00676-7
Huan Yue, Kaifeng Wu, Kanglin Liu, Luxia Gou, A. Huang, Hua Tang
{"title":"LINC02154 promotes the proliferation and metastasis of hepatocellular carcinoma by enhancing SPC24 promoter activity and activating the PI3K-AKT signaling pathway","authors":"Huan Yue, Kaifeng Wu, Kanglin Liu, Luxia Gou, A. Huang, Hua Tang","doi":"10.1007/s13402-022-00676-7","DOIUrl":"https://doi.org/10.1007/s13402-022-00676-7","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2022-05-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46341497","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: 14
FBXO32 targets PHPT1 for ubiquitination to regulate the growth of EGFR mutant lung cancer FBXO32靶向PHPT1进行泛素化以调节EGFR突变型癌症的生长
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2022-04-01 DOI: 10.1007/s13402-022-00669-6
Ning Zhang, Yifeng Liao, Weize Lv, Shunda Zhu, Yeqing Qiu, N. Chen, Mei Xiao, Hongyu Zhang
{"title":"FBXO32 targets PHPT1 for ubiquitination to regulate the growth of EGFR mutant lung cancer","authors":"Ning Zhang, Yifeng Liao, Weize Lv, Shunda Zhu, Yeqing Qiu, N. Chen, Mei Xiao, Hongyu Zhang","doi":"10.1007/s13402-022-00669-6","DOIUrl":"https://doi.org/10.1007/s13402-022-00669-6","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46155053","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: 6
Molecular signaling in pancreatic ductal metaplasia: emerging biomarkers for detection and intervention of early pancreatic cancer. 胰腺导管化生的分子信号传导:早期胰腺癌症检测和干预的新生物标志物
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2022-04-01 Epub Date: 2022-03-15 DOI: 10.1007/s13402-022-00664-x
Xiaojia Li, Jie He, Keping Xie

Pancreatic ductal metaplasia (PDM) is the transformation of potentially various types of cells in the pancreas into ductal or ductal-like cells, which eventually replace the existing differentiated somatic cell type(s). PDM is usually triggered by and manifests its ability to adapt to environmental stimuli and genetic insults. The development of PDM to atypical hyperplasia or dysplasia is an important risk factor for pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDA). Recent studies using genetically engineered mouse models, cell lineage tracing, single-cell sequencing and others have unraveled novel cellular and molecular insights in PDM formation and evolution. Those novel findings help better understand the cellular origins and functional significance of PDM and its regulation at cellular and molecular levels. Given that PDM represents the earliest pathological changes in PDA initiation and development, translational studies are beginning to define PDM-associated cell and molecular biomarkers that can be used to screen and detect early PDA and to enable its effective intervention, thereby truly and significantly reducing the dreadful mortality rate of PDA. This review will describe recent advances in the understanding of PDM biology with a focus on its underlying cellular and molecular mechanisms, and in biomarker discovery with clinical implications for the management of pancreatic regeneration and tumorigenesis.

胰腺导管增生(PDM)是指胰腺中潜在的各类细胞转变为导管或导管样细胞,并最终取代现有的分化体细胞类型。PDM 通常由环境刺激和遗传损伤引发,并表现出对环境刺激和遗传损伤的适应能力。PDM 发展为非典型增生或发育不良是胰腺上皮内瘤变(PanIN)和胰管腺癌(PDA)的重要风险因素。最近利用基因工程小鼠模型、细胞系追踪、单细胞测序等方法进行的研究揭示了 PDM 形成和进化过程中的细胞和分子新观点。这些新发现有助于更好地理解 PDM 的细胞起源、功能意义及其在细胞和分子水平上的调控。鉴于 PDM 代表了 PDA 发病和发展过程中最早的病理变化,转化研究正开始定义与 PDM 相关的细胞和分子生物标记物,这些标记物可用于筛选和检测早期 PDA 并进行有效干预,从而真正显著降低 PDA 的可怕死亡率。本综述将介绍对 PDM 生物学认识的最新进展,重点关注其潜在的细胞和分子机制,以及对胰腺再生和肿瘤发生管理具有临床意义的生物标志物的发现。
{"title":"Molecular signaling in pancreatic ductal metaplasia: emerging biomarkers for detection and intervention of early pancreatic cancer.","authors":"Xiaojia Li, Jie He, Keping Xie","doi":"10.1007/s13402-022-00664-x","DOIUrl":"10.1007/s13402-022-00664-x","url":null,"abstract":"<p><p>Pancreatic ductal metaplasia (PDM) is the transformation of potentially various types of cells in the pancreas into ductal or ductal-like cells, which eventually replace the existing differentiated somatic cell type(s). PDM is usually triggered by and manifests its ability to adapt to environmental stimuli and genetic insults. The development of PDM to atypical hyperplasia or dysplasia is an important risk factor for pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDA). Recent studies using genetically engineered mouse models, cell lineage tracing, single-cell sequencing and others have unraveled novel cellular and molecular insights in PDM formation and evolution. Those novel findings help better understand the cellular origins and functional significance of PDM and its regulation at cellular and molecular levels. Given that PDM represents the earliest pathological changes in PDA initiation and development, translational studies are beginning to define PDM-associated cell and molecular biomarkers that can be used to screen and detect early PDA and to enable its effective intervention, thereby truly and significantly reducing the dreadful mortality rate of PDA. This review will describe recent advances in the understanding of PDM biology with a focus on its underlying cellular and molecular mechanisms, and in biomarker discovery with clinical implications for the management of pancreatic regeneration and tumorigenesis.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2022-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47295483","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
BCL6 and the Notch pathway: a signaling axis leading to a novel druggable biotarget in triple negative breast cancer BCL6和Notch通路:三阴性乳腺癌中一个新的可药物生物靶点的信号传导轴
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2022-03-31 DOI: 10.1007/s13402-022-00663-y
F. De Santis, S. Romero-Cordoba, L. Castagnoli, T. Volpari, S. Faraci, G. Fucà, E. Tagliabue, F. de Braud, S. Pupa, M. Di Nicola
{"title":"BCL6 and the Notch pathway: a signaling axis leading to a novel druggable biotarget in triple negative breast cancer","authors":"F. De Santis, S. Romero-Cordoba, L. Castagnoli, T. Volpari, S. Faraci, G. Fucà, E. Tagliabue, F. de Braud, S. Pupa, M. Di Nicola","doi":"10.1007/s13402-022-00663-y","DOIUrl":"https://doi.org/10.1007/s13402-022-00663-y","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2022-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48399155","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: 6
Soluble TGFBI aggravates the malignancy of cholangiocarcinoma through activation of the ITGB1 dependent PPARγ signalling pathway 可溶性TGFBI通过激活ITGB1依赖性PPARγ信号通路加重胆管癌恶性程度
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2022-03-31 DOI: 10.1007/s13402-022-00668-7
Jungwhoi Lee, Jungsul Lee, Woogwang Sim, Jae-Hoon Kim
{"title":"Soluble TGFBI aggravates the malignancy of cholangiocarcinoma through activation of the ITGB1 dependent PPARγ signalling pathway","authors":"Jungwhoi Lee, Jungsul Lee, Woogwang Sim, Jae-Hoon Kim","doi":"10.1007/s13402-022-00668-7","DOIUrl":"https://doi.org/10.1007/s13402-022-00668-7","url":null,"abstract":"","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2022-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"44541301","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: 8
MAP3K1 expression is associated with progression and poor prognosis of hormone receptor-positive, HER2-negative early-stage breast cancer MAP3K1表达与激素受体阳性、her2阴性早期乳腺癌的进展和不良预后相关
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2020-08-06 DOI: 10.21203/rs.3.rs-53956/v1
S. Kuo, M. Wei, Yi-Hsuan Lee, Jui-Chueh Lin, Wen-Chi Yang, Shi‐Yi Yang, Chiun-Sheng Huang
Purpose In this study, we assessed whether the overexpression of MAP3K1 promotes the proliferation, migration, and invasion of breast cancer cells, which affect the prognosis of hormone receptor (HR)–positive, human epidermal growth factor receptor 2 (HER2)–negative early stage breast cancer. Methods Two HR-positive, HER2-negative breast cancer cell lines (MCF7 and T-47D) overexpressing MAP3K1 were transfected with two MAP3K1 short hairpin RNA plasmids (shMAP3K1 [#3] and shMAP3K1 [#5]). The proliferation, migration, and invasion of these cells were then examined. We assessed whether shMAP3K1 affects the cell cycle, levels of downstream signaling molecules (ERK, JNK, p38 MAPK, and NF-κB), and sensitivity to chemotherapeutic and hormonal agents. To assess the anti-tumor effect of MAP3K1 knockdown in the breast cancer orthotopic model, MCF7 and T-47D cells treated with or without shMAP3K1 (#3) and shMAP3K1 (#5) were inoculated into the mammary fat pads of mice. In total, 182 patients with HR-positive, HER2-negative T1 and T2 breast cancer and 0–3 nodal metastases were included. Additionally, 73 patients with T1 and T2 breast cancer and negative nodes who received adjuvant endocrine therapy alone were selected as an independent validation cohort. Results In both cell lines, shMAP3K1 (#3) and shMAP3K1 (#5) significantly reduced cell growth, migration, and invasion by downregulating MMP-9 and by blocking the G2/M phase of the cell cycle and its regulatory molecule cyclin B1. Moreover, both shMAP3K1 (#3) and shMAP3K1 (#5) downregulated ERK-, JNK-, p38 MAPK-, and NF-κB-dependent gene transcription and enhanced the sensitivity of both cell lines to doxorubicin, docetaxel, and tamoxifen. We observed that both shMAP3K1 (#3) and shMAP3K1 (#5) inhibited tumor growth compared with that in the scrambled group of MCF7 and T-47D cell orthotopic tumors. Patients with MAP3K1 overexpression exhibited significantly poorer 10-year disease-free survival (DFS) (70.4% vs. 88.6%, p  = 0.003) and overall survival (OS) (81.9% vs. 96.3%, p  = 0.001) than those without MAP3K1 overexpression. Furthermore, phospho-ERK ( p  < 0.001) and phospho-JNK ( p  < 0.001) expressions were significantly associated with MAP3K1 expression, and both phospho-ERK and phospho-JNK expressions were significantly correlated with poor 10-year DFS and OS. These biological findings, including a significant association between DFS and OS, and the expressions of MAP3K1, phospho-ERK, and phospho-JNK were further validated in an independent cohort. Multivariate analysis identified MAP3K1 expression as an independent poor prognostic factor for DFS and OS. Conclusion Our results indicate that the overexpression of MAP3K1 plays a major role in the poor prognosis of HR-positive, HER2-negative early stage breast cancer.
在本研究中,我们评估MAP3K1过表达是否促进乳腺癌细胞的增殖、迁移和侵袭,从而影响激素受体(HR)阳性、人表皮生长因子受体2 (HER2)阴性的早期乳腺癌的预后。方法用两种MAP3K1短发夹RNA质粒(shMAP3K1[#3]和shMAP3K1[#5])转染过表达MAP3K1的hr阳性和her2阴性乳腺癌细胞系MCF7和T-47D。然后检测这些细胞的增殖、迁移和侵袭。我们评估了shMAP3K1是否影响细胞周期、下游信号分子(ERK、JNK、p38 MAPK和NF-κB)的水平,以及对化疗和激素药物的敏感性。为了评估MAP3K1敲低在乳腺癌原位模型中的抗肿瘤作用,我们将经或不经shMAP3K1(#3)和shMAP3K1(#5)处理的MCF7和T-47D细胞接种于小鼠乳腺脂肪层。共纳入182例hr阳性、her2阴性的T1、T2乳腺癌及0-3淋巴结转移患者。另外,选择73例单独接受辅助内分泌治疗的T1和T2乳腺癌阴性淋巴结患者作为独立验证队列。结果在两种细胞系中,shMAP3K1(#3)和shMAP3K1(#5)通过下调MMP-9和阻断细胞周期的G2/M期及其调控分子细胞周期蛋白B1,显著降低细胞生长、迁移和侵袭。此外,shMAP3K1(#3)和shMAP3K1(#5)下调ERK-、JNK-、p38 MAPK-和NF-κ b依赖性基因转录,并增强两种细胞系对阿霉素、多西他赛和他莫昔芬的敏感性。我们观察到,与MCF7和T-47D细胞原位肿瘤的打乱组相比,shMAP3K1(#3)和shMAP3K1(#5)均抑制肿瘤生长。MAP3K1过表达患者的10年无病生存率(DFS) (70.4% vs. 88.6%, p = 0.003)和总生存率(OS) (81.9% vs. 96.3%, p = 0.001)明显低于无MAP3K1过表达患者。此外,phospho-ERK (p < 0.001)和phospho-JNK (p < 0.001)表达与MAP3K1表达显著相关,phospho-ERK和phospho-JNK表达与10年DFS和OS差显著相关。这些生物学发现,包括DFS和OS之间的显著关联,以及MAP3K1、phospho-ERK和phospho-JNK的表达,在一个独立的队列中得到了进一步的验证。多变量分析发现,MAP3K1表达是DFS和OS的独立不良预后因素。结论MAP3K1过表达在hr阳性、her2阴性的早期乳腺癌预后不良中起重要作用。
{"title":"MAP3K1 expression is associated with progression and poor prognosis of hormone receptor-positive, HER2-negative early-stage breast cancer","authors":"S. Kuo, M. Wei, Yi-Hsuan Lee, Jui-Chueh Lin, Wen-Chi Yang, Shi‐Yi Yang, Chiun-Sheng Huang","doi":"10.21203/rs.3.rs-53956/v1","DOIUrl":"https://doi.org/10.21203/rs.3.rs-53956/v1","url":null,"abstract":"Purpose In this study, we assessed whether the overexpression of MAP3K1 promotes the proliferation, migration, and invasion of breast cancer cells, which affect the prognosis of hormone receptor (HR)–positive, human epidermal growth factor receptor 2 (HER2)–negative early stage breast cancer. Methods Two HR-positive, HER2-negative breast cancer cell lines (MCF7 and T-47D) overexpressing MAP3K1 were transfected with two MAP3K1 short hairpin RNA plasmids (shMAP3K1 [#3] and shMAP3K1 [#5]). The proliferation, migration, and invasion of these cells were then examined. We assessed whether shMAP3K1 affects the cell cycle, levels of downstream signaling molecules (ERK, JNK, p38 MAPK, and NF-κB), and sensitivity to chemotherapeutic and hormonal agents. To assess the anti-tumor effect of MAP3K1 knockdown in the breast cancer orthotopic model, MCF7 and T-47D cells treated with or without shMAP3K1 (#3) and shMAP3K1 (#5) were inoculated into the mammary fat pads of mice. In total, 182 patients with HR-positive, HER2-negative T1 and T2 breast cancer and 0–3 nodal metastases were included. Additionally, 73 patients with T1 and T2 breast cancer and negative nodes who received adjuvant endocrine therapy alone were selected as an independent validation cohort. Results In both cell lines, shMAP3K1 (#3) and shMAP3K1 (#5) significantly reduced cell growth, migration, and invasion by downregulating MMP-9 and by blocking the G2/M phase of the cell cycle and its regulatory molecule cyclin B1. Moreover, both shMAP3K1 (#3) and shMAP3K1 (#5) downregulated ERK-, JNK-, p38 MAPK-, and NF-κB-dependent gene transcription and enhanced the sensitivity of both cell lines to doxorubicin, docetaxel, and tamoxifen. We observed that both shMAP3K1 (#3) and shMAP3K1 (#5) inhibited tumor growth compared with that in the scrambled group of MCF7 and T-47D cell orthotopic tumors. Patients with MAP3K1 overexpression exhibited significantly poorer 10-year disease-free survival (DFS) (70.4% vs. 88.6%, p  = 0.003) and overall survival (OS) (81.9% vs. 96.3%, p  = 0.001) than those without MAP3K1 overexpression. Furthermore, phospho-ERK ( p  < 0.001) and phospho-JNK ( p  < 0.001) expressions were significantly associated with MAP3K1 expression, and both phospho-ERK and phospho-JNK expressions were significantly correlated with poor 10-year DFS and OS. These biological findings, including a significant association between DFS and OS, and the expressions of MAP3K1, phospho-ERK, and phospho-JNK were further validated in an independent cohort. Multivariate analysis identified MAP3K1 expression as an independent poor prognostic factor for DFS and OS. Conclusion Our results indicate that the overexpression of MAP3K1 plays a major role in the poor prognosis of HR-positive, HER2-negative early stage breast cancer.","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2020-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45156513","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: 2
Piperlongumine potentiates the antitumor efficacy of oxaliplatin through ROS induction in gastric cancer cells. Piperlongumine通过ROS诱导增强奥沙利铂对癌症细胞的抗肿瘤作用
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2019-12-01 Epub Date: 2019-09-06 DOI: 10.1007/s13402-019-00471-x
Peichen Zhang, Lingyan Shi, Tingting Zhang, Lin Hong, Wei He, Peihai Cao, Xin Shen, Peisen Zheng, Yiqun Xia, Peng Zou

Purpose: Oxaliplatin is one of the most commonly used chemotherapeutic agents in the treatment of various cancers, including gastric cancer. It has, however, a narrow therapeutic index due to its toxicity and the occurrence of drug resistance. Therefore, there is a pressing need to develop novel therapies to potentiate the efficacy and reduce the toxicity of oxaliplatin. Piperlongumine (PL), an alkaloid isolated from Piper longum L., has recently been identified as a potent agent against cancer cells in vitro and in vivo. In the present study, we investigated whether PL can potentiate the antitumor effect of oxaliplatin in gastric cancer cells.

Methods: Cellular apoptosis and ROS levels were analyzed by flow cytometry. Thioredoxin reductase 1 (TrxR1) activity in gastric cancer cells or tumor tissues was determined using an endpoint insulin reduction assay. Western blotting was used to analyze the expression levels of the indicated proteins. Nude mice xenograft models were used to test the effects of PL and oxaliplatin combinations on gastric cancer cell growth in vivo.

Results: We found that PL significantly enhanced oxaliplatin-induced growth inhibition in both gastric and colon cancer cells. Moreover, we found that PL potentiated the antitumor effect of oxaliplatin by inhibiting TrxR1 activity. PL combined with oxaliplatin markedly suppressed the activity of TrxR1, resulting in the accumulation of ROS and, thereby, DNA damage induction and p38 and JNK signaling pathway activation. Pretreatment with antioxidant N-acetyl-L-cysteine (NAC) significantly abrogated the combined treatment-induced ROS generation, DNA damage and apoptosis. Importantly, we found that activation of the p38 and JNK signaling pathways prompted by PL and oxaliplatin was also reversed by NAC pretreatment. In vivo, we found that PL combined with oxaliplatin significantly suppressed tumor growth in a gastric cancer xenograft model, and effectively reduced the activity of TrxR1 in tumor tissues. Remarkably, we found that PL attenuated body weight loss evoked by oxaliplatin treatment.

Conclusions: Our data support a synergistic effect of PL and oxaliplatin and suggest that application of its combination may be more effective for the treatment of gastric cancer than oxaliplatin alone.

目的:奥沙利铂是治疗包括胃癌在内的各种癌症最常用的化疗药物之一。然而,由于其毒性和耐药性的出现,它的治疗指数较窄。因此,迫切需要开发新型疗法来增强奥沙利铂的疗效并降低其毒性。胡椒龙葵碱(Piperlongumine,PL)是从胡椒龙葵(Piper longum L.)中分离出来的一种生物碱,最近被确认为体外和体内抗癌细胞的有效药物。在本研究中,我们探讨了PL能否增强奥沙利铂对胃癌细胞的抗肿瘤作用:方法:采用流式细胞术分析细胞凋亡和 ROS 水平。采用终点胰岛素还原试验测定胃癌细胞或肿瘤组织中硫氧还原酶 1 (TrxR1) 的活性。用 Western 印迹法分析所述蛋白质的表达水平。裸鼠异种移植模型用于检测 PL 和奥沙利铂组合对胃癌细胞体内生长的影响:结果:我们发现 PL 能明显增强奥沙利铂诱导的胃癌和结肠癌细胞的生长抑制作用。此外,我们还发现 PL 通过抑制 TrxR1 的活性增强了奥沙利铂的抗肿瘤作用。PL与奥沙利铂联合使用可明显抑制TrxR1的活性,导致ROS积累,从而诱发DNA损伤,激活p38和JNK信号通路。使用抗氧化剂 N-乙酰-L-半胱氨酸(NAC)进行预处理可显著减少联合治疗诱导的 ROS 生成、DNA 损伤和细胞凋亡。重要的是,我们发现 NAC 预处理还能逆转 PL 和奥沙利铂对 p38 和 JNK 信号通路的激活。在体内,我们发现 PL 联合奥沙利铂能显著抑制胃癌异种移植模型中的肿瘤生长,并有效降低肿瘤组织中 TrxR1 的活性。值得注意的是,我们发现 PL 能减轻奥沙利铂治疗引起的体重下降:我们的数据支持 PL 和奥沙利铂的协同作用,并表明联合应用 PL 和奥沙利铂治疗胃癌可能比单独应用奥沙利铂更有效。
{"title":"Piperlongumine potentiates the antitumor efficacy of oxaliplatin through ROS induction in gastric cancer cells.","authors":"Peichen Zhang, Lingyan Shi, Tingting Zhang, Lin Hong, Wei He, Peihai Cao, Xin Shen, Peisen Zheng, Yiqun Xia, Peng Zou","doi":"10.1007/s13402-019-00471-x","DOIUrl":"10.1007/s13402-019-00471-x","url":null,"abstract":"<p><strong>Purpose: </strong>Oxaliplatin is one of the most commonly used chemotherapeutic agents in the treatment of various cancers, including gastric cancer. It has, however, a narrow therapeutic index due to its toxicity and the occurrence of drug resistance. Therefore, there is a pressing need to develop novel therapies to potentiate the efficacy and reduce the toxicity of oxaliplatin. Piperlongumine (PL), an alkaloid isolated from Piper longum L., has recently been identified as a potent agent against cancer cells in vitro and in vivo. In the present study, we investigated whether PL can potentiate the antitumor effect of oxaliplatin in gastric cancer cells.</p><p><strong>Methods: </strong>Cellular apoptosis and ROS levels were analyzed by flow cytometry. Thioredoxin reductase 1 (TrxR1) activity in gastric cancer cells or tumor tissues was determined using an endpoint insulin reduction assay. Western blotting was used to analyze the expression levels of the indicated proteins. Nude mice xenograft models were used to test the effects of PL and oxaliplatin combinations on gastric cancer cell growth in vivo.</p><p><strong>Results: </strong>We found that PL significantly enhanced oxaliplatin-induced growth inhibition in both gastric and colon cancer cells. Moreover, we found that PL potentiated the antitumor effect of oxaliplatin by inhibiting TrxR1 activity. PL combined with oxaliplatin markedly suppressed the activity of TrxR1, resulting in the accumulation of ROS and, thereby, DNA damage induction and p38 and JNK signaling pathway activation. Pretreatment with antioxidant N-acetyl-L-cysteine (NAC) significantly abrogated the combined treatment-induced ROS generation, DNA damage and apoptosis. Importantly, we found that activation of the p38 and JNK signaling pathways prompted by PL and oxaliplatin was also reversed by NAC pretreatment. In vivo, we found that PL combined with oxaliplatin significantly suppressed tumor growth in a gastric cancer xenograft model, and effectively reduced the activity of TrxR1 in tumor tissues. Remarkably, we found that PL attenuated body weight loss evoked by oxaliplatin treatment.</p><p><strong>Conclusions: </strong>Our data support a synergistic effect of PL and oxaliplatin and suggest that application of its combination may be more effective for the treatment of gastric cancer than oxaliplatin alone.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2019-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1007/s13402-019-00471-x","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"46083132","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: 40
Human bone marrow-derived mesenchymal stem cell-secreted exosomes overexpressing microRNA-34a ameliorate glioblastoma development via down-regulating MYCN. 过表达microRNA-34a的人骨髓间充质干细胞分泌外泌体通过下调MYCN改善胶质母细胞瘤的发展。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2019-12-01 Epub Date: 2019-07-22 DOI: 10.1007/s13402-019-00461-z
Bin Wang, Zhong-Hua Wu, Ping-Yang Lou, Chang Chai, Shuang-Yin Han, Jian-Fang Ning, Ming Li

Purpose: Exosomes play important roles in intercellular communication through signaling pathways affecting tumor microenvironment modulation and tumor proliferation, including those in glioblastoma (GBM). As yet, however, limited studies have been conducted on the inhibitory effect of human bone marrow-derived mesenchymal stem cell (hBMSC)-derived exosomes on GBM development. Therefore, we set out to assess the role of hBMSC secreted exosomes, in particular those carrying microRNA-34a (miR-34a), in the development of GBM.

Methods: Microarray-based expression analysis was employed to identify differentially expressed genes and to predict miRNAs regulating MYCN expression. Next, hBMSCs were transfected with a miR-34a mimic or inhibitor after which exosomes were isolated. Proliferation, apoptosis, migration, invasion and temozolomide (TMZ) chemosensitivity of exosome-exposed GBM cells (T-98G, LN229 and A-172) were measured in vitro. The mechanism underlying MYCN regulation was investigated using lentiviral transfections. The in vivo inhibitory effect of exosomal miR-34a was measured in nude mice xenografted with GBM cells through subcutaneous injection of hBMSCs with an upregulated miR34a content.

Results: We found that poorly-expressed miR-34a specifically targeted and negatively regulated the expression of MYCN in GBM cells. In addition we found that miR-34a was delivered to T-98G, LN229 and A-172 GBM cells via hBMSC-derived exosomes. Exogenous overexpression of miR-34a in hBMSC-derived exosomes resulted in inhibition of GBM cell proliferation, invasion, migration and tumorigenesis in vitro and in vivo, while promoting the chemosensitivity of GBM cells to TMZ by silencing MYCN.

Conclusions: From our data we conclude that hBMSC-derived exosomes overexpressing miR-34a may be instrumental for the therapeutic targeting and clinical management of GBM.

目的:外泌体通过影响肿瘤微环境调节和肿瘤增殖的信号通路在细胞间通讯中发挥重要作用,包括在胶质母细胞瘤(GBM)中。然而,迄今为止,关于人骨髓间充质干细胞(hBMSC)来源的外泌体对GBM发育的抑制作用的研究还很有限。因此,我们开始评估hBMSC分泌外泌体,特别是那些携带microRNA-34a (miR-34a)的外泌体在GBM发展中的作用。方法:采用微阵列表达分析鉴定差异表达基因,预测调节MYCN表达的mirna。接下来,用miR-34a模拟物或抑制剂转染hBMSCs,然后分离外泌体。体外检测外泌体暴露的GBM细胞(T-98G、LN229和A-172)的增殖、凋亡、迁移、侵袭及替莫唑胺(TMZ)的化学敏感性。利用慢病毒转染研究了MYCN调控的机制。通过皮下注射miR34a含量上调的hBMSCs,在移植GBM细胞的裸鼠中测量了外泌体miR-34a的体内抑制作用。结果:我们发现低表达的miR-34a特异性靶向并负向调节GBM细胞中MYCN的表达。此外,我们发现miR-34a通过hbmsc衍生的外泌体递送到T-98G、LN229和A-172 GBM细胞。在hbmsc来源的外泌体中外源性过表达miR-34a导致GBM细胞体外和体内增殖、侵袭、迁移和肿瘤发生受到抑制,同时通过沉默MYCN促进GBM细胞对TMZ的化学敏感性。结论:根据我们的数据,我们得出结论,hbmsc衍生的过表达miR-34a的外泌体可能有助于GBM的治疗靶向和临床管理。
{"title":"Human bone marrow-derived mesenchymal stem cell-secreted exosomes overexpressing microRNA-34a ameliorate glioblastoma development via down-regulating MYCN.","authors":"Bin Wang, Zhong-Hua Wu, Ping-Yang Lou, Chang Chai, Shuang-Yin Han, Jian-Fang Ning, Ming Li","doi":"10.1007/s13402-019-00461-z","DOIUrl":"https://doi.org/10.1007/s13402-019-00461-z","url":null,"abstract":"<p><strong>Purpose: </strong>Exosomes play important roles in intercellular communication through signaling pathways affecting tumor microenvironment modulation and tumor proliferation, including those in glioblastoma (GBM). As yet, however, limited studies have been conducted on the inhibitory effect of human bone marrow-derived mesenchymal stem cell (hBMSC)-derived exosomes on GBM development. Therefore, we set out to assess the role of hBMSC secreted exosomes, in particular those carrying microRNA-34a (miR-34a), in the development of GBM.</p><p><strong>Methods: </strong>Microarray-based expression analysis was employed to identify differentially expressed genes and to predict miRNAs regulating MYCN expression. Next, hBMSCs were transfected with a miR-34a mimic or inhibitor after which exosomes were isolated. Proliferation, apoptosis, migration, invasion and temozolomide (TMZ) chemosensitivity of exosome-exposed GBM cells (T-98G, LN229 and A-172) were measured in vitro. The mechanism underlying MYCN regulation was investigated using lentiviral transfections. The in vivo inhibitory effect of exosomal miR-34a was measured in nude mice xenografted with GBM cells through subcutaneous injection of hBMSCs with an upregulated miR34a content.</p><p><strong>Results: </strong>We found that poorly-expressed miR-34a specifically targeted and negatively regulated the expression of MYCN in GBM cells. In addition we found that miR-34a was delivered to T-98G, LN229 and A-172 GBM cells via hBMSC-derived exosomes. Exogenous overexpression of miR-34a in hBMSC-derived exosomes resulted in inhibition of GBM cell proliferation, invasion, migration and tumorigenesis in vitro and in vivo, while promoting the chemosensitivity of GBM cells to TMZ by silencing MYCN.</p><p><strong>Conclusions: </strong>From our data we conclude that hBMSC-derived exosomes overexpressing miR-34a may be instrumental for the therapeutic targeting and clinical management of GBM.</p>","PeriodicalId":9690,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2019-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1007/s13402-019-00461-z","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138476853","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: 26
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