Skip Navigation
Skip to contents

Cancer Res Treat : Cancer Research and Treatment

OPEN ACCESS

Search

Page Path
HOME > Search
17 "Proliferation"
Filter
Filter
Article category
Keywords
Publication year
Authors
Funded articles
Original Articles
Lung and Thoracic cancer
MLL4 Regulates the Progression of Non–Small-Cell Lung Cancer by Regulating the PI3K/AKT/SOX2 Axis
Yang Yang, Rongfang Qiu, Qiaoyou Weng, Ziwei Xu, Jingjing Song, Siyu Zhao, Miaomiao Meng, Dengke Zhang, Chunli Kong, Hailin Wang, Min Xu, Zhongwei Zhao, Jiansong Ji
Cancer Res Treat. 2023;55(3):778-803.   Published online January 26, 2023
DOI: https://doi.org/10.4143/crt.2022.1042
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
Mixed-lineage leukemia protein 4 (MLL4/KMT2D) is a histone methyltransferase, and its mutation has been reported to be associated with a poor prognosis in many cancers, including lung cancer. We investigated the function of MLL4 in lung carcinogenesis.
Materials and Methods
RNA sequencing (RNA-seq) in A549 cells transfected with control siRNA or MLL4 siRNA was performed. Also, we used EdU incorporation assay, colony formation assays, growth curve analysis, transwell invasion assays, immunohistochemical staining, and in vivo bioluminescence assay to investigate the function of MLL4 in lung carcinogenesis.
Results
We found that MLL4 expression was downregulated in non–small cell lung cancer (NSCLC) tissues compared to adjacent normal tissues and tended to decrease with disease stage progression. We analyzed the transcriptomes in control and MLL4- deficient cells using high-throughput RNA deep sequencing (RNA-seq) and identified a cohort of target genes, such as SOX2, ATF1, FOXP4, PIK3IP1, SIRT4, TENT5B, and LFNG, some of which are related to proliferation and metastasis. Our results showed that low expression of MLL4 promotes NSCLC cell proliferation and metastasis and is required for the maintenance of NSCLC stem cell properties.
Conclusion
Our findings identify an important role of MLL4 in lung carcinogenesis through transcriptional regulation of PIK3IP1, affecting the PI3K/AKT/SOX2 axis, and suggest that MLL4 could be a potential prognostic indicator and target for NSCLC therapy.

Citations

Citations to this article as recorded by  
  • Role and potential therapeutic value of histone methyltransferases in drug resistance mechanisms in lung cancer
    Linxiang Zhang, Xueying Zhang, Yan Shi, Yuhan Ni, Jiaojiao Fei, Zhixin Jin, Wenjuan Li, Xiaojing Wang, Nan Wu
    Frontiers in Oncology.2024;[Epub]     CrossRef
  • The multifaceted role of SOX2 in breast and lung cancer dynamics
    Kiavash Hushmandi, Seyed Hassan Saadat, Seyedalireza Mirilavasani, Salman Daneshi, Amir Reza Aref, Noushin Nabavi, Rasoul Raesi, Afshin Taheriazam, Mehrdad Hashemi
    Pathology - Research and Practice.2024; 260: 155386.     CrossRef
  • PIK3IP1: structure, aberration, function, and regulation in diseases
    Yingjie Jia, Pengxing He, Xubin Ma, Kaili Lv, Ying Liu, Yichao Xu
    European Journal of Pharmacology.2024; 977: 176753.     CrossRef
  • UBQLN4 promotes the proliferation and invasion of non-small cell lung cancer cell by regulating PI3K/AKT pathway
    Li He, Heng Chen, Bin Ruan, Li He, Ming Luo, Yulun Fu, Rui Zou
    Journal of Cancer Research and Clinical Oncology.2024;[Epub]     CrossRef
  • Role of histone methyltransferase KMT2D in BMSC osteogenesis via AKT signaling
    Zhichun Zhang, Yanyan Guo, Xuejun Gao, Xiaoyan Wang, Chanyuan Jin
    Regenerative Therapy.2024; 26: 775.     CrossRef
  • Landscape of targeted therapies for lung squamous cell carcinoma
    Qiuxuan Chen, Xiaoshuo Zheng, Weiting Cheng, Jian Li
    Frontiers in Oncology.2024;[Epub]     CrossRef
  • 4,440 View
  • 275 Download
  • 6 Web of Science
  • 6 Crossref
Close layer
G Protein–Coupled Receptor 30 Mediates the Anticancer Effects Induced by Eicosapentaenoic Acid in Ovarian Cancer Cells
Yue Zhao, Meng-Fei Zhao, Mei-Lin Yang, Tian-Yu Wu, Cong-Jian Xu, Jing-Mei Wang, Chao-Jun Li, Xi Li
Cancer Res Treat. 2020;52(3):815-829.   Published online March 5, 2020
DOI: https://doi.org/10.4143/crt.2019.380
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
While numerous epidemiological studies have indicated that omega-3 polyunsaturated fatty acids have anticancer properties in various cancers, the effects and mechanisms of eicosapentaenoic acid (EPA) in ovarian cancer cell growth are poorly understood.
Materials and Methods
ES2 ovarian clear cell carcinoma cells and SKOV3 adenocarcinoma cells were treated with palmitic acid or EPA, followed by flow cytometry and cell counting to measure apoptosis and proliferation, respectively. A modified protein lipid overlay assay was used to further verify whether EPA was a ligand of G protein–coupled receptor 30 (GPR30) in ES2 cells. The levels of apoptosis-related genes, phosphorylated AKT, and phosphorylated ERK1/2 were detected to explore the underlying mechanism. Finally, inhibitory effect of EPA on tumor growth via GPR30 was determined in vitro and in vivo.
Results
EPA suppressed ES2 ovarian clear cell carcinoma cells growth via GPR30, a novel EPA receptor, by inducing apoptosis. As a ligand of GPR30, EPA activated the GPR30-cAMP– protein kinase A signaling pathway. When GPR30 was suppressed by siRNA or its inhibitor G15, the antiproliferative action of EPA was impaired. Furthermore, EPA inhibited tumor growth by blocking the activation of AKT and ERK. In the mouse xenograft model, EPA decreased tumor volume and weight through GPR30 by blocking tumor cell proliferation.
Conclusion
These results confirm that EPA is a tumor suppressor in human ovarian clear cell carcinoma cells and functions through a novel fatty acid receptor, GPR30, indicating a mechanistic linkage between omega-3 fatty acids and cancers.

Citations

Citations to this article as recorded by  
  • Integrative analysis of proteomics and lipidomic profiles reveal the fat deposition and meat quality in Duroc × Guangdong small spotted pig
    Zhuosui Wu, Zhonggang Wang, Pan Wang, Leiyan Cheng, Jianhao Li, Yanfeng Luo, Linfang Yang, Linfeng Li, Jianhua Zeng, Bin Hu
    Frontiers in Veterinary Science.2024;[Epub]     CrossRef
  • The G Protein-Coupled Estrogen Receptor GPER in the Development and Progression of Cancer
    Liliana Torres-López, Miguel Olivas-Aguirre, Oxana Dobrovinskaya
    Receptors.2024; 3(2): 220.     CrossRef
  • ACSL4-Mediated Membrane Phospholipid Remodeling Induces Integrin β1 Activation to Facilitate Triple-Negative Breast Cancer Metastasis
    Yuxiang Qiu, Xing Wang, Yan Sun, Ting Jin, Rui Tang, Xinyue Zhou, Ming Xu, Yubi Gan, Rui Wang, Haojun Luo, Manran Liu, Xi Tang
    Cancer Research.2024; 84(11): 1856.     CrossRef
  • The Role of Estrogen across Multiple Disease Mechanisms
    Xiuting Xiang, Praneetha Palasuberniam, Rahmawati Pare
    Current Issues in Molecular Biology.2024; 46(8): 8170.     CrossRef
  • Role of Estrogen Receptor β, G-Protein Coupled Estrogen Receptor and Estrogen-Related Receptors in Endometrial and Ovarian Cancer
    Susanne Schüler-Toprak, Maciej Skrzypczak, Carsten Gründker, Olaf Ortmann, Oliver Treeck
    Cancers.2023; 15(10): 2845.     CrossRef
  • Divergent Metabolic Effects of Metformin Merge to Enhance Eicosapentaenoic Acid Metabolism and Inhibit Ovarian Cancer In Vivo
    Mary P. Udumula, Laila M. Poisson, Indrani Dutta, Nivedita Tiwari, Seongho Kim, Jasdeep Chinna-Shankar, Ghassan Allo, Sharif Sakr, Miriana Hijaz, Adnan R. Munkarah, Shailendra Giri, Ramandeep Rattan
    Cancers.2022; 14(6): 1504.     CrossRef
  • Functional metabolomics revealed functional metabolic-characteristics of chronic hepatitis that is significantly differentiated from acute hepatitis in mice
    Tianyu Wang, Longlong Hu, Jiongjiong Lu, Mengqing Xiao, Jinging Liu, Huiyu Xia, Haitao Lu
    Pharmacological Research.2022; 180: 106248.     CrossRef
  • Effects of Omega-3 Intake on Adolescent Health
    Xiuyuan Zhang
    Highlights in Science, Engineering and Technology.2022; 19: 89.     CrossRef
  • Determination of lipid profiles of Dezhou donkey meat using an LC‐MS‐based lipidomics method
    Mengmeng Li, Mingxia Zhu, Wenqiong Chai, Yonghui Wang, Dongmei Fan, Mengqing Lv, Xiaojing Jiang, Yongxiang Liu, Qingxin Wei, Changfa Wang
    Journal of Food Science.2021; 86(10): 4511.     CrossRef
  • Connexins and cAMP Cross-Talk in Cancer Progression and Metastasis
    Chang-Xu Chen, Kai-Jun Luo, Jia-Peng Yang, Yun-Chao Huang, Eduardo R. Cardenas, Bruce J. Nicholson, Jean X. Jiang
    Cancers.2020; 13(1): 58.     CrossRef
  • 8,531 View
  • 179 Download
  • 8 Web of Science
  • 10 Crossref
Close layer
Low Doses of Nonylphenol Promote Growth of Colon Cancer Cells through Activation of ERK1/2 via G Protein‒Coupled Receptor 30
Ming Xie, Jin-Long Liang, Han-Dong Huang, Mai-Jian Wang, Tao Zhang, Xue-Feng Yang
Cancer Res Treat. 2019;51(4):1620-1631.   Published online May 15, 2019
DOI: https://doi.org/10.4143/crt.2018.340
AbstractAbstract PDFPubReaderePub
Purpose
Nonylphenol (NP) is an endocrine disruptor found in products such as cleaners, plastics, and detergents. It exerts actions similar to endogenous 17β-estradiol (E2) and is reported to influence various cancers. However, its role in colon cancer remains elusive.
Materials and Methods
Colon cancer cell lines COLO 205 and SW480 were employed in our study. The cells were treated with NP or E2 followed by measurement of apoptosis and proliferation using flow cytometry and MTT assays, respectively. G protein–coupled estrogen receptor 30 (GPR30) expression was visualized using immunofluorescence and Western blot. To investigate the underlying mechanism, the expression levels of GPR30, p-protein kinase A (PKA), c-myc, cyclin D1, and ERK1/2 were analyzed using Western blot. Meanwhile, the GPR30 antagonist G15 was utilized to validate the role of GPR30 in colon cancer progression. Finally, the effect of a GPR30 inhibitor on tumor growth was determined in vivo using tumor xenograft mouse models.
Results
NP facilitated the proliferation of colon cancer cells and induced apoptosis failure in vitro. Western blot revealed increased GPR30 expression levels in response to NP treatment. Cyclin D1, p-PKA, c-myc, and proliferating cell nuclear antigen, proteins that regulate the cell cycle, were all upregulated by NP, and NP-mediated ERK1/2 activation and subsequent cell proliferation were abrogated by the GPR30 inhibitor G15. Moreover, colon cancer mice that received G15 administration demonstrated impaired tumor growth in vivo.
Conclusion
Low dose NP promotes the growth of colon tumors through GPR30-mediated activation of ERK1/2 signaling.

Citations

Citations to this article as recorded by  
  • Role of sex steroids in colorectal cancer: pathomechanisms and medical applications
    Jianglan Wu
    American Journal of Cancer Research.2024; 14(7): 3200.     CrossRef
  • 17β-estradiol in colorectal cancer: friend or foe?
    Zihong Wu, Chong Xiao, Jiamei Wang, Min Zhou, Fengming You, Xueke Li
    Cell Communication and Signaling.2024;[Epub]     CrossRef
  • Magnetic nanoparticles for eliminating endocrine-disrupting compounds in water treatment – a quantitative systematic analysis
    Juliana Guimarães, Igor Taveira, Thuane Mendes Anacleto, Alex Enrich-Prast, Fernanda Abreu
    Frontiers in Environmental Science.2024;[Epub]     CrossRef
  • Implications of estrogen and its receptors in colorectal carcinoma
    Plabon Kumar Das, Joti Saha, Suja Pillai, Alfred K.‐Y. Lam, Vinod Gopalan, Farhadul Islam
    Cancer Medicine.2023; 12(4): 4367.     CrossRef
  • Endocrine-Disrupting Chemicals and Disease Endpoints
    Changhwan Ahn, Eui-Bae Jeung
    International Journal of Molecular Sciences.2023; 24(6): 5342.     CrossRef
  • Hsp27, a potential EcR target, protects nonylphenol-induced cellular and organismal toxicity in Drosophila melanogaster
    Shiwangi Dwivedi, Leonard Clinton D'Souza, Nidhi Ganesh Shetty, Shamprasad Varija Raghu, Anurag Sharma
    Environmental Pollution.2022; 293: 118484.     CrossRef
  • Effect of nonylphenol on the colonic mucosa in rats and intervention with zinc-selenium green tea (Camellia sinensis)
    Shixu Li, Mucong Zheng, Xuefeng Yang, Jianling Zhang, Jie Xu, Jie Yu
    Toxicology Research.2022; 11(1): 122.     CrossRef
  • Nonylphenol regulates TL1A through the AhR/HDAC2/HNF4α pathway in endothelial cells to promote the angiogenesis of colorectal cancer
    Tao Zhang, Wei-Wei Ning, Jie Zhang, Fu-Jian Xu, Xing-Qin Wang, Zheng-Biao Li, Ming Xie
    Toxicology and Applied Pharmacology.2022; 436: 115854.     CrossRef
  • Effects of subchronic exposure of nonylphenol on the expression of immune-related factors and estrogen receptors in the spleen of rats
    Xiangjun Fu, Jie Xu, Chengyu Ni, Degang Yu, Haibo Wang, Pan Wang, Man Luo, Jie Yu
    Environmental Sciences Europe.2022;[Epub]     CrossRef
  • Mechanism of nonylphenol induced gastric inflammation through NF-κB/NLRP3 signaling pathway
    Jie Xu, Shixu Li, Xuefeng Yang, Haibo Wang, Lina Ma, Yuan Shen, Jie Yu
    Toxicology.2022; 479: 153294.     CrossRef
  • Nonylphenol Promoted Epithelial–Mesenchymal Transition in Colorectal Cancer Cells by Upregulating the Expression of Regulator of Cell Cycle
    Nian-jie Zhang, Yuanwei Zhang, Shuo Yin, Du-ji Ruan, Nian He, Xu Chen, Xue-feng Yang
    Chemical Research in Toxicology.2022; 35(9): 1533.     CrossRef
  • Co-exposure to BPA and DEHP enhances susceptibility of mammary tumors via up-regulating Esr1/HDAC6 pathway in female rats
    Xuan Zhang, Cheng Cheng, Guopei Zhang, Mingyang Xiao, Liuli Li, Shengwen Wu, Xiaobo Lu
    Ecotoxicology and Environmental Safety.2021; 221: 112453.     CrossRef
  • Coiled-Coil Domain Containing 80 Suppresses Nonylphenol-Induced Colorectal Cancer Cell Proliferation by Inhibiting the Activation of ERK1/2
    Jing Wang, Yuan-wei Zhang, Nian-jie Zhang, Shuo Yin, Du-ji Ruan, Nian He, Xu Chen, Xue-feng Yang
    Frontiers in Cell and Developmental Biology.2021;[Epub]     CrossRef
  • Residential proximity to industrial pollution sources and colorectal cancer risk: A multicase-control study (MCC-Spain)
    Javier García-Pérez, Nerea Fernández de Larrea-Baz, Virginia Lope, Antonio J. Molina, Cristina O'Callaghan-Gordo, María Henar Alonso, Marta María Rodríguez-Suárez, Benito Mirón-Pozo, Juan Alguacil, Inés Gómez-Acebo, Nieves Ascunce, Mercedes Vanaclocha-Esp
    Environment International.2020; 144: 106055.     CrossRef
  • 6,689 View
  • 174 Download
  • 15 Web of Science
  • 14 Crossref
Close layer
Cathepsin C Interacts with TNF-α/p38 MAPK Signaling Pathway to Promote Proliferation and Metastasis in Hepatocellular Carcinoma
Guo-Pei Zhang, Xiao Yue, Shao-Qiang Li
Cancer Res Treat. 2020;52(1):10-23.   Published online April 26, 2019
DOI: https://doi.org/10.4143/crt.2019.145
AbstractAbstract PDFPubReaderePub
Purpose
Although cathepsin C (CTSC) has been reported to maintain malignant biological properties in various cancers, its functions in hepatocellular carcinoma (HCC) remain obscure. We aimed to investigate the potential role of CTSC in HCC.
Materials and Methods
HCC tissue microarrays (n=122) were employed to analyze the correlation between CTSC expression and clinicopathological characteristics through immunohistochemistry staining. Quantitative real-time polymerase chain reaction, western blot assay, Cell Counting Kit-8 assay, colony formation, cell migration, and invasion assays, xenograft mice model were adopted to validate what had been indicated by the bioinformatic web tools.
Results
By bioinformatic tools and tissue microarrays, CTSC was found upregulated in HCC compared with normal liver tissues, and its higher expression was correlated with poor prognosis of HCC patients (hazard ratio, 2.402; 95% confidence interval, 1.493 to 3.865; p < 0.001). By gain/loss-of-function assays, we implicated that CTSC functioned as an oncogene to promote the proliferation and metastasis of HCC cells. Mechanistically, we revealed that CTSC was involved in several cancer-related signaling pathways by Gene Set Enrichment Analysis, among which tumor necrosis factor α (TNF-α)/p38 pathway was verified to be activated by CTSC. Furthermore, we found that TNF-α could activate CTSC expression in a concentration- dependent manner. Ralimetinib, an oral p38 mitogen-activated protein kinase (MAPK) inhibitor could inhibit CTSC expression. These indicated a potential positive feedback loop between CTSC and TNF-α/MAPK (p38) signaling.
Conclusion
Taken together, CTSC plays an important role in the growth and metastasis of HCC and may be a promising therapeutic target upon HCC.

Citations

Citations to this article as recorded by  
  • CTSC promoted the migration and invasion of glioma cells via activation of STAT3/SERPINA3 axis
    Qi Li, Chenguang Wan, Zhifei Zhang, Guangwei Liu, Song Wang
    Gene.2024; 893: 147948.     CrossRef
  • Characterization and function analysis of cathepsin C in Marsupenaeus japonicus
    Zuhao Tu, Jian Zhong, Hongquan Li, Liangjuan Sun, Yucong Huang, Shiping Yang, Yishan Lu, Shuanghu Cai
    Fish & Shellfish Immunology.2024; 146: 109379.     CrossRef
  • Cathepsin B: The dawn of tumor therapy
    Fuxian Liu, Ting Zhou, Shangzu Zhang, Yangyang Li, Yan Chen, Zhiming Miao, Xin Wang, Gengqiang Yang, Qiyang Li, Liying Zhang, Yongqi Liu
    European Journal of Medicinal Chemistry.2024; 269: 116329.     CrossRef
  • Targeting inflammation as cancer therapy
    Manni Wang, Siyuan Chen, Xuemei He, Yong Yuan, Xiawei Wei
    Journal of Hematology & Oncology.2024;[Epub]     CrossRef
  • DNA Damage-driven Inflammatory Cytokines: Reprogramming of Tumor Immune Microenvironment and Application of Oncotherapy
    Meng-jie Wang, Yu Xia, Qing-lei Gao
    Current Medical Science.2024; 44(2): 261.     CrossRef
  • Identification of PANoptosis-related subtypes, construction of a prognosis signature, and tumor microenvironment landscape of hepatocellular carcinoma using bioinformatic analysis and experimental verification
    Guoqing Ouyang, Qiuyun Li, Yangnian Wei, Wenbin Dai, Haojian Deng, Youli Liu, Jiaguang Li, Mingjuan Li, Shunwen Luo, Shuang Li, Yunying Liang, Guandong Pan, Jianqing Yang, Tao Gan
    Frontiers in Immunology.2024;[Epub]     CrossRef
  • Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury
    Jing Yu, Yu Fu, Jiameng Gao, Qingqing Zhang, Nan Zhang, Zhiyuan Zhang, Xuemei Jiang, Chang Chen, Zongmei Wen
    Redox Biology.2024; 74: 103231.     CrossRef
  • Protective Effects of Hydroalcoholic Leaf Extract of Origanum Vulgare against Paraquat-induced Lung Toxicity in Rats: The Role of the Inflammation-related Markers and Oxidative Stress
    Ali Sharifi-Rigi, Esfandiar Heidarian, Fatemeh Zal
    The Natural Products Journal.2024;[Epub]     CrossRef
  • Cathepsin C regulates tumor progression via the Yes-associated protein signaling pathway in non-small cell lung cancer
    Nayoung Kim
    American Journal of Cancer Research.2024; 14(1): 97.     CrossRef
  • DOX-PLGA Nanoparticles Effectively Suppressed the Expression of Pro-Inflammatory Cytokines TNF-a, IL-6, iNOS, and IL-1β in MCF-7 Breast Cancer Cell Line
    Rawan Hassan Al-Saeedi, Mohammad Khalaj-Kondori, Mohammad Ali Hosseinpour Feizi, Jafar Hajavi
    Reports of Biochemistry and Molecular Biology.2024; 12(4): 530.     CrossRef
  • Cathepsin C in health and disease: from structural insights to therapeutic prospects
    Chakriya Chitsamankhun, Nutwara Siritongtaworn, B. P. J. Fournier, Kanokwan Sriwattanapong, Thanakorn Theerapanon, Lakshman Samaranayake, Thantrira Porntaveetus
    Journal of Translational Medicine.2024;[Epub]     CrossRef
  • Cathepsin C Promotes Tumorigenesis in Bladder Cancer by Activating the Wnt/β-catenin Signalling Pathway
    Xinsheng Wang, Yong Jia, Dawen Wang
    Frontiers in Bioscience-Landmark.2024;[Epub]     CrossRef
  • Cysteine cathepsins: From diagnosis to targeted therapy of cancer
    Ana Ercegovič Rot, Matija Hrovatin, Bor Bokalj, Ernestina Lavrih, Boris Turk
    Biochimie.2024; 226: 10.     CrossRef
  • Searching for novel cellular targets for MASLD and HCC within the humble lysosomal cathepsins
    Alejandro del Castillo-Cruz, Maria Fernández-Fernández, Anna Moles
    Exploration of Digestive Diseases.2024; 3(6): 428.     CrossRef
  • Exploring the potential targets of the Abrus cantoniensis Hance in the treatment of hepatitis E based on network pharmacology
    Ziheng Xu, Can Wang, Zuxiang Luan, Dapei Zhang, Baiqing Dong
    Frontiers in Veterinary Science.2023;[Epub]     CrossRef
  • Characterization, Potential Prognostic Value, and Immune Heterogeneity of Cathepsin C in Diffuse Glioma
    Quanwei Zhou, Shasha Li, Xuejun Yan, Hecheng Zhu, Weidong Liu, Youwei Guo, Hongjuan Xu, Wen Yin, Xuewen Li, Qian Yang, Hui Liu, Xingjun Jiang, Caiping Ren
    Current Bioinformatics.2023; 18(1): 76.     CrossRef
  • Correlation Analysis of CTSB Promoter Polymorphism and Function in Patients with Dilated Cardiomyopathy
    Yu Zhou, Shuang Gao, Liangcai Ding, Han Yan, Shuchao Pang, Bo Yan
    DNA and Cell Biology.2023; 42(4): 203.     CrossRef
  • The role of cytokines in the pathogenesis of malignant neoplasms
    V. L. Rybkina, G. V. Adamova, D. S. Oslina
    Сибирский научный медицинский журнал.2023; 43(2): 15.     CrossRef
  • The role of lysosomal peptidases in glioma immune escape: underlying mechanisms and therapeutic strategies
    Hao Liu, Jie Peng, Linzhen Huang, Dong Ruan, Yuguang Li, Fan Yuan, Zewei Tu, Kai Huang, Xingen Zhu
    Frontiers in Immunology.2023;[Epub]     CrossRef
  • Cathepsin C: structure, function, and pharmacological targeting
    Milena Stojkovska Docevska, Marko Novinec
    Rare Disease and Orphan Drugs Journal.2023;[Epub]     CrossRef
  • Longitudinal Serum Proteomics Characterization of CD19-CAR-T Cell Therapy for B-Cell Malignancies
    Youming Wang, Rui Sun, Weigang Ge, Lei Xue, Qianwen Xu, Hui Xu, Sujun Li, Miaomiao Wu, Tiannan Guo, Xingbing Wang
    Journal of Proteome Research.2023; 22(9): 2985.     CrossRef
  • The Role of Cysteine Protease Cathepsins B, H, C, and X/Z in Neurodegenerative Diseases and Cancer
    Veronika Stoka, Olga Vasiljeva, Hiroshi Nakanishi, Vito Turk
    International Journal of Molecular Sciences.2023; 24(21): 15613.     CrossRef
  • The Role of Cytokines in the Pathogenesis of Malignant Neoplasms
    V. L. Rybkina, G. V. Adamova, D. S. Oslina
    Cell and Tissue Biology.2023; 17(6): 608.     CrossRef
  • Dipeptidyl peptidase 1 inhibition as a potential therapeutic approach in neutrophil-mediated inflammatory disease
    James D. Chalmers, Ralph Kettritz, Brice Korkmaz
    Frontiers in Immunology.2023;[Epub]     CrossRef
  • Nanoparticle-based drug delivery systems in cancer: A focus on inflammatory pathways
    Amir R. Afshari, Mehdi Sanati, Hamid Mollazadeh, Prashant Kesharwani, Thomas P. Johnston, Amirhossein Sahebkar
    Seminars in Cancer Biology.2022; 86: 860.     CrossRef
  • Cysteine cathepsin C: a novel potential biomarker for the diagnosis and prognosis of glioma
    Xingbo Cheng, Zhishuai Ren, Zhendong Liu, Xiang Sun, Rongjun Qian, Chen Cao, Binfeng Liu, Jialin Wang, Hongbo Wang, Yuqi Guo, Yanzheng Gao
    Cancer Cell International.2022;[Epub]     CrossRef
  • Lysosomal peptidases—intriguing roles in cancer progression and neurodegeneration
    Janko Kos, Ana Mitrović, Milica Perišić Nanut, Anja Pišlar
    FEBS Open Bio.2022; 12(4): 708.     CrossRef
  • Regulatory T-cells-related signature for identifying a prognostic subtype of hepatocellular carcinoma with an exhausted tumor microenvironment
    Genhao Zhang
    Frontiers in Immunology.2022;[Epub]     CrossRef
  • Recent progress in molecular mechanisms of postoperative recurrence and metastasis of hepatocellular carcinoma
    Zhao-Shan Niu, Wen-Hong Wang, Xiao-Jun Niu
    World Journal of Gastroenterology.2022; 28(46): 6433.     CrossRef
  • Construction and validation of prognostic signature for hepatocellular carcinoma basing on hepatitis B virus related specific genes
    Lei Wang, Manman Qiu, Lili Wu, Zexing Li, Xinyi Meng, Lu He, Bing Yang
    Infectious Agents and Cancer.2022;[Epub]     CrossRef
  • Identification of new potential antigen recognized by γδT cells in hepatocellular carcinoma
    Xueyan Xi, Yang Guo, Min Zhu, Fen Qiu, Feifei Lei, Gang Li, Boyu Du
    Cancer Immunology, Immunotherapy.2021; 70(7): 1917.     CrossRef
  • Inflammatory Cytokines in Cancer: Comprehensive Understanding and Clinical Progress in Gene Therapy
    Tianxia Lan, Li Chen, Xiawei Wei
    Cells.2021; 10(1): 100.     CrossRef
  • Harnessing Tumor Necrosis Factor Alpha to Achieve Effective Cancer Immunotherapy
    María Florencia Mercogliano, Sofía Bruni, Florencia Mauro, Patricia Virginia Elizalde, Roxana Schillaci
    Cancers.2021; 13(3): 564.     CrossRef
  • Dual Role of TNF and LTα in Carcinogenesis as Implicated by Studies in Mice
    Ekaterina O. Gubernatorova, Almina I. Polinova, Mikhail M. Petropavlovskiy, Olga A. Namakanova, Alexandra D. Medvedovskaya, Ruslan V. Zvartsev, Georgij B. Telegin, Marina S. Drutskaya, Sergei A. Nedospasov
    Cancers.2021; 13(8): 1775.     CrossRef
  • Resveratrol inhibited the metastatic behaviors of cisplatin‐resistant human oral cancer cells via phosphorylation of ERK/p‐38 and suppression of MMP‐2/9
    Wen‐Shin Chang, Chia‐Wen Tsai, Jai‐Sing Yang, Yuan‐Man Hsu, Liang‐Chun Shih, Hong‐Yi Chiu, Da‐Tian Bau, Fuu‐Jen Tsai
    Journal of Food Biochemistry.2021;[Epub]     CrossRef
  • Identifying Dendritic Cell–Related Genes Through a Co-Expression Network to Construct a 12-Gene Risk-Scoring Model for Predicting Hepatocellular Carcinoma Prognosis
    Chaoyuan Huang, Xiaotao Jiang, Yuancheng Huang, Lina Zhao, Peiwu Li, Fengbin Liu
    Frontiers in Molecular Biosciences.2021;[Epub]     CrossRef
  • Tumor-Induced Inflammatory Cytokines and the Emerging Diagnostic Devices for Cancer Detection and Prognosis
    Apriliana E. R. Kartikasari, Cesar S. Huertas, Arnan Mitchell, Magdalena Plebanski
    Frontiers in Oncology.2021;[Epub]     CrossRef
  • The multifaceted role of cathepsins in liver disease
    Paloma Ruiz-Blázquez, Valeria Pistorio, María Fernández-Fernández, Anna Moles
    Journal of Hepatology.2021; 75(5): 1192.     CrossRef
  • Aging-related genes are potential prognostic biomarkers for patients with gliomas
    Gelei Xiao, Xiangyang Zhang, Xun Zhang, Yuanbing Chen, Zhiwei Xia, Hui Cao, Jun Huang, Quan Cheng
    Aging.2021; 13(9): 13239.     CrossRef
  • Cathepsin C inhibitors as anti-inflammatory drug discovery: Challenges and opportunities
    Xiao Bao Shen, Xing Chen, Zhao Yan Zhang, Fu Fang Wu, Xin Hua Liu
    European Journal of Medicinal Chemistry.2021; 225: 113818.     CrossRef
  • Cathepsin C Is Involved in Macrophage M1 Polarization via p38/MAPK Pathway in Sudden Cardiac Death
    Jialin Dai, Jiangjin Liu, Qiong Zhang, Yang An, Bing Xia, Changwu Wan, Yuanyuan Zhang, Yanni Yu, Jie Wang, Hangang Yu
    Cardiovascular Therapeutics.2021; 2021: 1.     CrossRef
  • Cathepsin C inhibition as a potential treatment strategy in cancer
    Brice Korkmaz, Anne-Sophie Lamort, Roxane Domain, Céline Beauvillain, Artur Gieldon, Ali Önder Yildirim, Georgios T. Stathopoulos, Moez Rhimi, Dieter E. Jenne, Ralph Kettritz
    Biochemical Pharmacology.2021; 194: 114803.     CrossRef
  • Papillary Thyroid Carcinoma Landscape and Its Immunological Link With Hashimoto Thyroiditis at Single-Cell Resolution
    Jun Pan, Fang Ye, Chengxuan Yu, Qinsheng Zhu, Jiaqi Li, Yaohui Zhang, Hedi Tian, Yunjin Yao, Minjie Zhu, Yibin Shen, Feng Zhu, Yingying Wang, Xinhui Zhou, Guoji Guo, Yijun Wu
    Frontiers in Cell and Developmental Biology.2021;[Epub]     CrossRef
  • In Silico Comparative Studies of Tumor Necrosis Factor Alpha-Induced Proteins
    Sri Venkata Anandavalli Nallapati#, Manasa Reddy Sanaga#, Swapna Aedhu#, Naga Harini Parimi, Shravan Kumar Gunda
    International Journal Of Pharmaceutical And Phytopharmacological Research.2021; 11(6): 21.     CrossRef
  • Praeruptorin B Mitigates the Metastatic Ability of Human Renal Carcinoma Cells through Targeting CTSC and CTSV Expression
    Chia-Liang Lin, Tung-Wei Hung, Tsung-Ho Ying, Chi-Jui Lin, Yi-Hsien Hsieh, Chien-Min Chen
    International Journal of Molecular Sciences.2020; 21(8): 2919.     CrossRef
  • 10,003 View
  • 407 Download
  • 46 Web of Science
  • 45 Crossref
Close layer
Paip1 Indicated Poor Prognosis in Cervical Cancer and Promoted Cervical Carcinogenesis
Nan Li, Junjie Piao, Xinyue Wang, Ki-Yeol Kim, Jung Yoon Bae, Xiangshan Ren, Zhenhua Lin
Cancer Res Treat. 2019;51(4):1653-1665.   Published online April 19, 2019
DOI: https://doi.org/10.4143/crt.2018.544
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
This study was aimed to investigate the role of poly(A)-binding protein-interacting protein 1 (Paip1) in cervical carcinogenesis.
Materials and Methods
The expression of Paip1 in normal cervical epithelial tissues and cervical cancer (CC) tissues were detected by immunohistochemistry. In vivo and in vitro assays were performed to validate effect of Paip1 on CC progression.
Results
Paip1 was found to be up-regulated in CC, which was linked with shorter survival. Knockdown of Paip1 inhibited cell growth, induced apoptosis and cell cycle arrest in CC cells, whereas its overexpression reversed these effects. The in vivo tumor model confirmed the pro-tumor role of Paip1 in CC growth.
Conclusion
Altogether, the investigation demonstrated the clinical significance of Paip1 expression, which prompted that the up-regulated of Paip1 can presumably be a potential prognostic and progression marker for CC.

Citations

Citations to this article as recorded by  
  • Prioritizing cervical cancer candidate genes using chaos game and fractal-based time series approach
    T. Mallikarjuna, N. B. Thummadi, Vaibhav Vindal, P. Manimaran
    Theory in Biosciences.2024; 143(3): 183.     CrossRef
  • Knockdown of PAIP1 Inhibits Breast Cancer Cell Proliferation by Regulating Cyclin E2 mRNA Stability
    Wenqing Yang, Qingkun Wang, Qi Li, Yue Han, Yu Zhang, Lu Zhu, Lianhua Zhu, Junjie Piao
    Molecular Carcinogenesis.2024;[Epub]     CrossRef
  • Human Proteome Microarray identifies autoantibodies to tumor‐associated antigens as serological biomarkers for the diagnosis of hepatocellular carcinoma
    Qian Yang, Hua Ye, Guiying Sun, Keyan Wang, Liping Dai, Cuipeng Qiu, Jianxiang Shi, Jicun Zhu, Xiao Wang, Peng Wang
    Molecular Oncology.2023; 17(5): 887.     CrossRef
  • PAIP1 regulates expression of immune and inflammatory response associated genes at transcript level in liver cancer cell
    Jianfeng Zheng, Weiwei Fan, Xiaoyu Zhang, Weili Quan, Yunfei Wu, Mengni Shu, Moyang Chen, Ming Liang
    PeerJ.2023; 11: e15070.     CrossRef
  • Advances in HPV‐associated tumor management: Therapeutic strategies and emerging insights
    Xiangjin Gong, Hao Chi, Zhijia Xia, Guanhu Yang, Gang Tian
    Journal of Medical Virology.2023;[Epub]     CrossRef
  • Prioritizing the candidate genes related to cervical cancer using the moment of inertia tensor
    Neelesh Babu Thummadi, Mallikarjuna T., Vaibhav Vindal, Manimaran P.
    Proteins: Structure, Function, and Bioinformatics.2022; 90(2): 363.     CrossRef
  • Effect of PAIP1 on the metastatic potential and prognostic significance in oral squamous cell carcinoma
    Neeti Swarup, Kyoung-Ok Hong, Kunal Chawla, Su-Jung Choi, Ji-Ae Shin, Kyu-Young Oh, Hye-Jung Yoon, Jae-Il Lee, Sung-Dae Cho, Seong-Doo Hong
    International Journal of Oral Science.2022;[Epub]     CrossRef
  • CRISPR/Cas9 Screening for Identification of Genes Required for the Growth of Ovarian Clear Cell Carcinoma Cells
    Ayako Kawabata, Tomoatsu Hayashi, Yoko Akasu-Nagayoshi, Ai Yamada, Naomi Shimizu, Naoko Yokota, Ryuichiro Nakato, Katsuhiko Shirahige, Aikou Okamoto, Tetsu Akiyama
    Current Issues in Molecular Biology.2022; 44(4): 1587.     CrossRef
  • Circ_0005576 Exerts an Oncogenic Role in Cervical Cancer via miR-1305-Dependent Regulation of PAIP1
    Yajing Wang, Fang Du, Zongyuan Xie, Junhao Lai, Yuanjie Li, Yongping Xu, Rui Tong
    Reproductive Sciences.2022; 29(9): 2647.     CrossRef
  • The p140Cap adaptor protein as a molecular hub to block cancer aggressiveness
    Vincenzo Salemme, Costanza Angelini, Jennifer Chapelle, Giorgia Centonze, Dora Natalini, Alessandro Morellato, Daniela Taverna, Emilia Turco, Ugo Ala, Paola Defilippi
    Cellular and Molecular Life Sciences.2021; 78(4): 1355.     CrossRef
  • Effects of inducing apoptosis and inhibiting proliferation of siRNA on polyadenylate‐binding protein‐interacting protein 1 in tongue cell carcinoma
    Huixu Xie, Lisa Yang, Qin Hu, Yingqi Song, Xiaoyi Wang, Liming Zhou, Longjiang Li
    Head & Neck.2020; 42(12): 3623.     CrossRef
  • Inhibition of carnitine palmitoyl transferase 1A-induced fatty acid oxidation suppresses cell progression in gastric cancer
    Liqiang Wang, Changfeng Li, Yumei Song, ZhenKun Yan
    Archives of Biochemistry and Biophysics.2020; 696: 108664.     CrossRef
  • Whole-genome resequencing of Dulong Chicken reveal signatures of selection
    Q. Wang, D. Li, A. Guo, M. Li, L. Li, J. Zhou, S. K. Mishra, G. Li, Y. Duan, Q. Li
    British Poultry Science.2020; 61(6): 624.     CrossRef
  • 8,229 View
  • 214 Download
  • 13 Web of Science
  • 13 Crossref
Close layer
Jab1 Silencing Inhibits Proliferation and Sensitizes to Cisplatin in Biliary Tract Cancer
Ah-Rong Nam, Ji-Won Kim, Ji Eun Park, Ju-Hee Bang, Mei Hua Jin, Do-Youn Oh, Yung-Jue Bang
Cancer Res Treat. 2019;51(3):886-900.   Published online October 1, 2018
DOI: https://doi.org/10.4143/crt.2018.375
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
Jab1 is a coactivator of c-Jun that enhances the transcriptional function of c-Jun. Jab1 is frequently overexpressed in various cancers and is associatedwith poor prognosis of cancer patients. Thus, Jab1 could be a potential therapeutic target in cancer. However, the role of Jab1 in biliary tract cancer (BTC) has not been studied.
Materials and Methods
We performed in vitro and in vivo experiments to evaluate the therapeutic potential ofJab1 inhibition in BTC.
Results
Among 8 BTC cell lines, many showed higher Jab1 expression levels. In addition, Jab1 silencing by siRNA increased p27 expression levels. SNU478 and HuCCT-1 cells exhibited profound Jab1 knockdown and increased p27 expression by Jab1-specific siRNA transfection. Jab1 silencing induced anti-proliferative and anti-migratory effects and resulted in G1 cell cycle arrest in SNU478 and HuCCT-1 cells. In addition, Jab1 silencing potentiated the anti-proliferative and anti-migratory effects of cisplatin by increasing DNA damage. Interestingly,Jab1 knockdown increased PTEN protein half-life, resulting in increased PTEN expression. In the HuCCT-1 mouse xenograft model, stable knockdown of Jab1 by shRNA also showed anti-proliferative effects in vivo, with decreased Ki-67 expression and AKT phosphorylation and increased Terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling and p27 expression.
Conclusion
Jab1 knockdown demonstrated anti-proliferative and anti-migratory effects in BTC cells by increasing DNA damage and stabilizing PTEN, resulting in G1 cell cycle arrest. In addition, Jab1 silencing potentiated the anti-proliferative effects of cisplatin. Our data suggest that Jab1 may be a potential therapeutic target in BTC that is worthy of further investigations.

Citations

Citations to this article as recorded by  
  • COP9 signalosome complex is a prognostic biomarker and corresponds with immune infiltration in hepatocellular carcinoma
    Jiahui Liu, Dexing Han, Junfeng Xuan, Jinye Xie, Weijia Wang, Quan Zhou, Kang Chen
    Aging.2024; 16(6): 5264.     CrossRef
  • Pan-cancer analyses of Jab1/COPS5 reveal oncogenic role and clinical outcome in human cancer
    Liping Wang, Xiaojiao Zeng, Gui Yang, Guohong Liu, Yunbao Pan
    Heliyon.2022; 8(12): e12553.     CrossRef
  • Jab1/Cops5: a promising target for cancer diagnosis and therapy
    Chunjue Yuan, Dong Wang, Guohong Liu, Yunbao Pan
    International Journal of Clinical Oncology.2021; 26(7): 1159.     CrossRef
  • 8,482 View
  • 225 Download
  • 6 Web of Science
  • 3 Crossref
Close layer
MicroRNA-373 Inhibits Cell Proliferation and Invasion via Targeting BRF2 in Human Non-small Cell Lung Cancer A549 Cell Line
Lei Wang, Junfeng Qu, Li Zhou, Fei Liao, Ju Wang
Cancer Res Treat. 2018;50(3):936-949.   Published online October 12, 2017
DOI: https://doi.org/10.4143/crt.2017.302
AbstractAbstract PDFPubReaderePub
Purpose
The purpose of this study was to investigate the biological role and mechanism of miR-373 targeting of TFIIB-related factor 2 (BRF2) in the regulation of non-small cell lung cancer (NSCLC) cells. Materials­and­Methods miRNA microarray chip analysis of four paired NSCLC and adjacent non-tumor tissues was performed. Quantitative real-time polymerase chain reaction (qRT-PCR) andwestern blotting were used to detect the expression levels of miR-373 and BRF2 in NSCLC tissues and cell lines. The dual-luciferase reporter method was performed to determine if BRF2 is a target of miR-373. MTT, wound-healing, Transwell, and flow cytometric assays were conducted to examine the proliferation, migration, invasion, and cell cycle progression of NSCLC A549 cells, respectively; western blotting was used to detect the expression of epithelial-mesenchymal transition (EMT)–related proteins.
Results
The miRNA microarray chip analysis demonstrated that miR-373 was down-regulated in NSCLC tissues, and this result was confirmed by qRT-PCR. Additionally, miR-373 was confirmed to target BRF2. Moreover, miR-373 expression was inversely correlated with BRF2 expression in NSCLC tissues and cell lines; both miR-373 down-regulation and BRF2 up-regulation were strongly associated with the clinicopathological features and prognosis of NSCLC patients. In vitro, overexpression of miR-373 markedly inhibited cell proliferation, migration, and invasion; up-regulated the expression of E-cadherin; and down-regulated the expression of N-cadherin and Snail in A549 cell. Knockdown BRF2 by siRNA resulted in effects similar to those caused by overexpression of miR-373.
Conclusion
MiR-373 is decreased in NSCLC, and overexpression of miR-373 can suppress cell EMT, and inhibit the proliferation, migration, and invasion of NSCLC A549 cells by targeting BRF2.

Citations

Citations to this article as recorded by  
  • MicroRNA‑373‑3p inhibits the proliferation and invasion of non‑small‑cell lung cancer cells by targeting the GAB2/PI3K/AKT pathway
    Xunxia Zhu, Xiaoyu Chen, Xuelin Zhang, Liting Zhao, Xiaoyong Shen
    Oncology Letters.2024;[Epub]     CrossRef
  • The role of EMG1 in lung adenocarcinoma progression: Implications for prognosis and immune cell infiltration
    Xingwei Wu, Zhenguo Wu, Zehang Xie, Haoyu Huang, Yingying Wang, Kun Lv, Hui Yang, Xiaocen Liu
    International Immunopharmacology.2024; 138: 112553.     CrossRef
  • The functions and molecular mechanisms of Tribbles homolog 3 (TRIB3) implicated in the pathophysiology of cancer
    Anam Arif, Ameer A. Alameri, Umer Bin Tariq, Shakeel Ahmed Ansari, Hader Ibrahim Sakr, Maytham T. Qasim, Fadhil F.M. Aljoborae, Andrés Alexis Ramírez-Coronel, Hijran Sanaan Jabbar, Gamal A. Gabr, Rasoul Mirzaei, Sajad Karampoor
    International Immunopharmacology.2023; 114: 109581.     CrossRef
  • Characteristics and Prognosis of 8p11.23-Amplified Squamous Lung Carcinomas
    Ioannis A. Voutsadakis
    Journal of Clinical Medicine.2023; 12(5): 1711.     CrossRef
  • MicroRNAs in cancer metastasis: biological and therapeutic implications
    Marie C. Sell, Charmaine A. Ramlogan-Steel, Jason C. Steel, Bijay P. Dhungel
    Expert Reviews in Molecular Medicine.2023;[Epub]     CrossRef
  • MALAT1/ mir-1-3p mediated BRF2 expression promotes HCC progression via inhibiting the LKB1/AMPK signaling pathway
    Guang-Zhen Li, Guang-Xiao Meng, Guo-Qiang Pan, Xiao Zhang, Lun-Jie Yan, Rui-Zhe Li, Zi-Niu Ding, Si-Yu Tan, Dong-Xu Wang, Bao-wen Tian, Yu-Chuan Yan, Zhao-Ru Dong, Jian-Guo Hong, Tao Li
    Cancer Cell International.2023;[Epub]     CrossRef
  • Processing body (P-body) and its mediators in cancer
    Bernard Nsengimana, Faiz Ali Khan, Ebenezeri Erasto Ngowi, Xuefeng Zhou, Yu Jin, Yuting Jia, Wenqiang Wei, Shaoping Ji
    Molecular and Cellular Biochemistry.2022; 477(4): 1217.     CrossRef
  • MicroRNAs, Tristetraprolin Family Members and HuR: A Complex Interplay Controlling Cancer-Related Processes
    Cyril Sobolewski, Laurent Dubuquoy, Noémie Legrand
    Cancers.2022; 14(14): 3516.     CrossRef
  • Amplification of 8p11.23 in cancers and the role of amplicon genes
    Ioannis A. Voutsadakis
    Life Sciences.2021; 264: 118729.     CrossRef
  • MicroRNAs and Lung Cancer: A Review Focused on Targeted Genes
    Yao-Hui Wang, Zhi-Ruo Zhu, De Tong, Rui Zhou, Kui Xiao, Ling Peng
    Exploratory Research and Hypothesis in Medicine.2021; 000(000): 1.     CrossRef
  • MiR-373 Inhibits the Epithelial-Mesenchymal Transition of Prostatic Cancer via Targeting Runt-Related Transcription Factor 2
    Jianyi Pang, Limei Dai, Chen Zhang, Qinglei Zhang, Enas Abdulhay
    Journal of Healthcare Engineering.2021; 2021: 1.     CrossRef
  • miR-96-5p is the tumor suppressor in osteosarcoma via targeting SYK
    Taiping Wang, Yong Xu, Xin Liu, Yong Zeng, Lei Liu
    Biochemical and Biophysical Research Communications.2021; 572: 49.     CrossRef
  • MicroRNA as an Important Target for Anticancer Drug Development
    Zhiwen Fu, Liu Wang, Shijun Li, Fen Chen, Kathy Ka-Wai Au-Yeung, Chen Shi
    Frontiers in Pharmacology.2021;[Epub]     CrossRef
  • lncRNA SNHG16 promotes glioma tumorigenicity through miR-373/EGFR axis by activating PI3K/AKT pathway
    Xiang-Yang Zhou, Hong Liu, Zheng-Bin Ding, Hai-Peng Xi, Guang-Wei Wang
    Genomics.2020; 112(1): 1021.     CrossRef
  • Molecular characterization of lung cancer: A two‐miRNA prognostic signature based on cancer stem‐like cells related genes
    Yanping Cheng, Sheng Yang, Bo Shen, Yan Zhang, Xiaomei Zhang, Tong Liu, Siyi Xu, Jing Sui, Lihong Yin, Yuepu Pu, Geyu Liang
    Journal of Cellular Biochemistry.2020; 121(4): 2889.     CrossRef
  • miR-93, miR-373, and miR-17-5p Negatively Regulate the Expression of TBP2 in Lung Cancer
    Ye Li, Min Liang, Yunhui Zhang, Bing Yuan, Wenchao Gao, Zhizhou Shi, Jie Bai
    Frontiers in Oncology.2020;[Epub]     CrossRef
  • BRF2 as a promising indicator for radical lymph-node dissection surgery in patients with cN0 squamous cell carcinoma of the middle thoracic esophagus
    Yu Tian, Cong Wang, Ming Lu
    Surgery Today.2019; 49(2): 158.     CrossRef
  • Long noncoding RNA MNX1‐AS1 contributes to lung cancer progression through the miR‐527/BRF2 pathway
    Haibo Liu, Leng Han, Zhengjia Liu, Nan Gao
    Journal of Cellular Physiology.2019; 234(8): 13843.     CrossRef
  • MiRNA-195-5p Functions as a Tumor Suppressor and a Predictive of Poor Prognosis in Non-small Cell Lung Cancer by Directly Targeting CIAPIN1
    Jing Zheng, Tingting Xu, Feng Chen, Ying Zhang
    Pathology & Oncology Research.2019; 25(3): 1181.     CrossRef
  • Amyloid precursor protein and its phosphorylated form in non-small cell lung carcinoma
    Shigehiro Ito, Yasuhiro Miki, Ryoko Saito, Chihiro Inoue, Yoshinori Okada, Hironobu Sasano
    Pathology - Research and Practice.2019; 215(8): 152463.     CrossRef
  • An Inventive Report of Inducing Apoptosis in Non-Small Cell Lung Cancer (NSCLC) Cell Lines by Transfection of MiR-4301
    Abbas J. Avval, Ahmad Majd, Naghmeh Gholipour, Kambiz A. Noghabi, Anna Ohradanova-Repic, Ghasem Ahangari
    Anti-Cancer Agents in Medicinal Chemistry.2019; 19(13): 1609.     CrossRef
  • miR‑802 inhibits the aggressive behaviors of non‑small cell lung cancer cells by directly targeting FGFR1
    Jiexia Zhang, Jun Li, Shiyue Li, Chengzhi Zhou, Yinyin Qin, Xiaoxiang Li
    International Journal of Oncology.2019;[Epub]     CrossRef
  • Development and characterisation of a panel of phosphatidylinositide 3-kinase – mammalian target of rapamycin inhibitor resistant lung cancer cell lines
    Susan Heavey, Paul Dowling, Gillian Moore, Martin P. Barr, Niamh Kelly, Stephen G. Maher, Sinead Cuffe, Stephen P. Finn, Kenneth J. O’Byrne, Kathy Gately
    Scientific Reports.2018;[Epub]     CrossRef
  • 9,766 View
  • 350 Download
  • 25 Web of Science
  • 23 Crossref
Close layer
Downregulation of HuR Inhibits the Progression of Esophageal Cancer through Interleukin-18
Xiaohui Xu, Cheng Song, Zhihua Chen, Chenxiao Yu, Yi Wang, Yiting Tang, Judong Luo
Cancer Res Treat. 2018;50(1):71-87.   Published online February 24, 2017
DOI: https://doi.org/10.4143/crt.2017.013
AbstractAbstract PDFSupplementary MaterialPubReaderePub
Purpose
The purpose of this study was to investigate the effect of human antigen R (HuR) downregulation and the potential target genes of HuR on the progression of esophageal squamous cell carcinoma (ESCC).
Materials and Methods
In this study, a proteomics assay was used to detect the expression of proteins after HuR downregulation, and a luciferase assay was used to detect the potential presence of a HuR binding site on the 3’-untranslated region (3'-UTR) of interleukin 18 (IL-18). In addition, colony formation assay, MTT, EdU incorporation assay, Western blot, flow cytometry, immunohistochemistry, transwell invasion assay, and wound healing assay were used.
Results
In the present study, we found that the expression of both HuR protein and mRNA levels were higher in tumor tissues than in the adjacent tissues. HuR downregulation significantly suppressed cell proliferation. In addition, the metastasis of esophageal cancer cells was inhibited, while the expression of E-cadherin was increased and the expression of matrix metalloproteinase (MMP) 2, MMP9, and vimentin was decreased after HuR knockdown. Moreover, silencing of HuR disturbed the cell cycle of ESCC cells mainly by inducing G1 arrest. Furthermore, proteomics analysis showed that downregulation of HuR in TE-1 cells resulted in 100 upregulated and 122 downregulated proteins, including IL-18 as a significantly upregulated protein. The expression of IL-18 was inversely regulated by HuR. IL-18 expression was decreased in ESCC tissues, and exogenous IL-18 significantly inhibited the proliferation and metastasis of ESCC cells. The 3'-UTR of IL-18 harbored a HuR binding site, as shown by an in vitro luciferase assay.
Conclusion
HuR plays an important role in the progression of esophageal carcinoma by targeting IL-18, which may be a potential therapeutic target for the treatment of ESCC.

Citations

Citations to this article as recorded by  
  • Mechanistic insights into the antitumoral potential and in vivo antiproliferative efficacy of a silver-based core@shell nanosystem
    Guillermo Aragoneses-Cazorla, Roberto Alvarez-Fernandez Garcia, Angelica Martinez-Lopez, Milagros Gomez Gomez, Maria Vallet-Regí, Sonia Castillo-Lluva, Blanca González, Jose L. Luque-Garcia
    International Journal of Pharmaceutics.2024; 655: 124023.     CrossRef
  • Blocking SLC7A11 attenuates the proliferation of esophageal squamous cell carcinoma cells
    Wen-Ting Li, Xin Jin, Sheng-Jie Song, Chong Wang, Chuang Fu, Wen Jiang, Jie Bai, Zhi-Zhou Shi
    Animal Cells and Systems.2024; 28(1): 237.     CrossRef
  • Editorial: Radiotherapy for esophageal cancer: Molecular mechanism and novel targets to improve radiosensitivity
    Yanqing Liu, Adam Yongxin Ye, Jordan Lu, Jiaxing Yang, Xiao Zheng, Xiaodong Li
    Frontiers in Oncology.2023;[Epub]     CrossRef
  • HuR Affects the Radiosensitivity of Esophageal Cancer by Regulating the EMT-Related Protein Snail
    Yan Hu, Qing Li, Ke Yi, Chi Yang, Qingjun Lei, Guanghui Wang, Qianyun Wang, Xiaohui Xu
    Frontiers in Oncology.2022;[Epub]     CrossRef
  • Identification of a Novel Pyroptosis-Related Gene Signature Indicative of Disease Prognosis and Treatment Response in Skin Cutaneous Melanoma
    An-An Li, Yu Zhang, Wei-Lai Tong, Jiang-Wei Chen, Shan-Hu Huang, Jia-Ming Liu, Zhi-Li Liu
    International Journal of General Medicine.2022; Volume 15: 6145.     CrossRef
  • MicroRNAs, Tristetraprolin Family Members and HuR: A Complex Interplay Controlling Cancer-Related Processes
    Cyril Sobolewski, Laurent Dubuquoy, Noémie Legrand
    Cancers.2022; 14(14): 3516.     CrossRef
  • The role of human antigen R (HuR) in modulating proliferation, senescence and radiosensitivity of skin cells
    Daojiang Yu, Yahui Feng, Zhiqiang Jiang, Tao Yan, Kai Fang, Yuhong Shi, Jie Zhang, Shuyu Zhang
    Experimental and Therapeutic Medicine.2022;[Epub]     CrossRef
  • IFI35 is involved in the regulation of the radiosensitivity of colorectal cancer cells
    Yan Hu, Bing Wang, Ke Yi, Qingjun Lei, Guanghui Wang, Xiaohui Xu
    Cancer Cell International.2021;[Epub]     CrossRef
  • LncRNA BBOX1‐AS1 upregulates HOXC6 expression through miR‐361‐3p and HuR to drive cervical cancer progression
    Jun Xu, Baohua Yang, Lifeng Wang, Yunheng Zhu, Xiuxiang Zhu, Ziyin Xia, Zhen Zhao, Ling Xu
    Cell Proliferation.2020;[Epub]     CrossRef
  • Angustoline Inhibited Esophageal Tumors Through Regulating LKB1/AMPK/ELAVL1/LPACT2 Pathway and Phospholipid Remodeling
    Huiying Li, Cheng Zhang, Min Zhang, Qianqian Yao, Huaigu Yang, Linlin Fan, Nan Zheng
    Frontiers in Oncology.2020;[Epub]     CrossRef
  • The RNA-Binding Protein HuR in Digestive System Tumors
    Xiaoqing Song, Xin Shi, Wenjuan Li, Fa Zhang, Zhonglin Cai
    BioMed Research International.2020; 2020: 1.     CrossRef
  • Proteogenomic examination of esophageal squamous cell carcinoma (ESCC): new lines of inquiry
    Shobha Dagamajalu, Manavalan Vijayakumar, Rohan Shetty, D. A. B. Rex, Chinmaya Narayana Kotimoole, T. S. Keshava Prasad
    Expert Review of Proteomics.2020; 17(9): 649.     CrossRef
  • Interleukin-18 Is a Prognostic Marker and Plays a Tumor Suppressive Role in Colon Cancer
    Xiaodong Feng, Zhijun Zhang, Peng Sun, Guanghui Song, Lu Wang, Zhenqing Sun, Ning Yuan, Qing Wang, Limin Lun, Roberta Rizzo
    Disease Markers.2020; 2020: 1.     CrossRef
  • Proteomics in Drug Development: The Dawn of a New Era?
    Maria Frantzi, Agnieszka Latosinska, Harald Mischak
    PROTEOMICS – Clinical Applications.2019;[Epub]     CrossRef
  • IL-33 in the tumor microenvironment is associated with the accumulation of FoxP3-positive regulatory T cells in human esophageal carcinomas
    Guanglin Cui, Zhenfeng Li, Jingli Ren, Aping Yuan
    Virchows Archiv.2019; 475(5): 579.     CrossRef
  • The Multifaceted Roles of Pyroptotic Cell Death Pathways in Cancer
    Man Wang, Shuai Jiang, Yinfeng Zhang, Peifeng Li, Kun Wang
    Cancers.2019; 11(9): 1313.     CrossRef
  • Activation of ferritinophagy is required for the RNA-binding protein ELAVL1/HuR to regulate ferroptosis in hepatic stellate cells
    Zili Zhang, Zhen Yao, Ling Wang, Hai Ding, Jiangjuan Shao, Anping Chen, Feng Zhang, Shizhong Zheng
    Autophagy.2018; 14(12): 2083.     CrossRef
  • Deficiency of IL-18 Aggravates Esophageal Carcinoma Through Inhibiting IFN-γ Production by CD8+T Cells and NK Cells
    Jiantao Li, Gang Qiu, Baoshuan Fang, Xiaohui Dai, Jianhui Cai
    Inflammation.2018; 41(2): 667.     CrossRef
  • 11,713 View
  • 299 Download
  • 22 Web of Science
  • 18 Crossref
Close layer
Effects and Mechanisms of Metformin on the Proliferation of Esophageal Cancer Cells In Vitro and In Vivo
Jian-Cai Tang, Rui An, Yi-Qing Jiang, Jian Yang
Cancer Res Treat. 2017;49(3):778-789.   Published online November 11, 2016
DOI: https://doi.org/10.4143/crt.2015.485
AbstractAbstract PDFPubReaderePub
Purpose
The purpose of this study was to observe the effects of metformin on human esophageal cancer cell and to investigate its possible mechanisms.
Materials and Methods
Cell viability was detected by using a Cell Counting Kit-8, while cell cycle and apoptosis were assessed by flow cytometry and western blot was used to measure the expression of the related proteins. RNAi was used to knockout pyruvate kinase muscle isozyme 2 (PKM2). An Eca109 tumor model was established to evaluate the antitumor effect in vivo. Immunohistochemistry was determined based on the expression of PKM2 and Bim in tumor tissues. Tunnel was used to assess tumor cell apoptosis.
Results
Esophageal cancer cells viability was reduced after metformin treatment. The cell cycle was arrested in the G0/G1 phase, apoptosis was induced, caspase 3 was activated, caspase 9 was downregulated, and the pro-apoptotic protein Bim increased. Further study revealed that metformin could suppress the expression of insulin-like growth factor 1 receptor and its downstream proteins, phosphoinositide 3-kinase (PI3K), protein kinase B (AKT/PKB), phosphorylation of AKT (pAKT), mammalian target of rapamycin (mTOR), p70S6K, and PKM2. Insulin-like growth factor 1 partly reversed metfromin-induced apoptosis and attenuated the repression effect of metfomin to PI3K, pAKT, and PKM2. Knockout PKM2 resulted in the activation of caspase 3, down-regulation of caspase 9, and increased expression of Bim. In the Eca109 xenograft model, metformin significantly reduced tumor growth. Furthermore, we found that metformin treatment increased the rate of apoptosis, down-regulation of PKM2, and up-regulation of Bim in tumor tissues.
Conclusion
Metformin restrained esophageal cancer cell proliferation partly by suppressing the PI3K/AKT/mTOR pathway.

Citations

Citations to this article as recorded by  
  • Metformin in Esophageal Carcinoma: Exploring Molecular Mechanisms and Therapeutic Insights
    Stavros P. Papadakos, Alexandra Argyrou, Vasileios Lekakis, Konstantinos Arvanitakis, Polyxeni Kalisperati, Ioanna E. Stergiou, Ippokratis Konstantinidis, Dimitrios Schizas, Theocharis Koufakis, Georgios Germanidis, Stamatios Theocharis
    International Journal of Molecular Sciences.2024; 25(5): 2978.     CrossRef
  • Ianus Bifrons: The Two Faces of Metformin
    Umberto Goglia, Iderina Hasballa, Claudia Teti, Mara Boschetti, Diego Ferone, Manuela Albertelli
    Cancers.2024; 16(7): 1287.     CrossRef
  • Metformin and the PI3K/AKT signaling pathway: implications for cancer, cardiovascular, and central nervous system diseases
    Hemin Ashayeri Ahmadabad, Somayeh Mohammadi Panah, Hojat Ghasemnejad‐Berenji, Shabnam Ghojavand, Morteza Ghasemnejad-Berenji, Mohammad Rafi Khezri
    Naunyn-Schmiedeberg's Archives of Pharmacology.2024;[Epub]     CrossRef
  • Changes in the expression of cancer- and metastasis-related genes and proteins after metformin treatment under different metabolic conditions in endometrial cancer cells
    Carsten Lange, Jana Brüggemann, Theresa Thüner, Julia Jauckus, Thomas Strowitzki, Ariane Germeyer
    Heliyon.2023; 9(6): e16678.     CrossRef
  • Metformin Induces a Caspase 3-Unrelated Apoptosis in Human Colorectal Cancer Cell Lines HCT116 and SW620
    Bustanur Rosidi, Diana Priyatno, Teguh Pribadi Putra, Irawan Yusuf
    Cancer Management and Research.2023; Volume 15: 475.     CrossRef
  • Metformin and Cancer Hallmarks: Molecular Mechanisms in Thyroid, Prostate and Head and Neck Cancer Models
    Mirian Galliote Morale, Rodrigo Esaki Tamura, Ileana Gabriela Sanchez Rubio
    Biomolecules.2022; 12(3): 357.     CrossRef
  • Metformin-Induced Heat Shock Protein Family A Member 6 Is a Promising Biomarker of Esophageal Squamous Cell Carcinoma
    Nobufumi Sekino, Masayuki Kano, Sohei Kobayashi, Kentaro Murakami, Haruhito Sakata, Takeshi Toyozumi, Satoshi Endo, Yasunori Matsumoto, Hiroshi Suito, Masahiko Takahashi, Ryota Otsuka, Masaya Yokoyama, Tadashi Shiraishi, Koichiro Okada, Toshiki Kamata, Ta
    Oncology.2022; 100(5): 267.     CrossRef
  • The Role of PKM2 in the Regulation of Mitochondrial Function: Focus on Mitochondrial Metabolism, Oxidative Stress, Dynamic, and Apoptosis. PKM2 in Mitochondrial Function
    Jing Gao, Yuwei Zhao, Tao Li, Xueqi Gan, Haiyang Yu, Daniel Lopez Malo
    Oxidative Medicine and Cellular Longevity.2022; 2022: 1.     CrossRef
  • Association of Common Use Pharmaceuticals in Reducing Risk of Esophageal Adenocarcinoma: A SEER–Medicare Analysis
    Holli A. Loomans-Kropp, Matthew Chaloux, Ellen Richmond, Asad Umar
    Cancer Prevention Research.2021; 14(2): 195.     CrossRef
  • Advances and challenges in the treatment of esophageal cancer
    Shiming He, Jian Xu, Xiujun Liu, Yongsu Zhen
    Acta Pharmaceutica Sinica B.2021; 11(11): 3379.     CrossRef
  • Changes in protein expression due to metformin treatment and hyperinsulinemia in a human endometrial cancer cell line
    Carsten Lange, Amanda Machado Weber, Ronny Schmidt, Christoph Schroeder, Thomas Strowitzki, Ariane Germeyer, Jae-Wook Jeong
    PLOS ONE.2021; 16(3): e0248103.     CrossRef
  • Glucose and fatty acid metabolism involved in the protective effect of metformin against ulipristal-induced endometrial changes in rats
    Marwa S. Hamza, Eman Ramadan, Salama A. Salama
    Scientific Reports.2021;[Epub]     CrossRef
  • 12-Epi-Napelline Inhibits Leukemia Cell Proliferation via the PI3K/AKT Signaling Pathway In Vitro and In Vivo
    Jia Han, Wei Hou, Bi-qing Cai, Fan Zhang, Jian-cai Tang, Xing Li
    Evidence-Based Complementary and Alternative Medicine.2021; 2021: 1.     CrossRef
  • Photodynamic therapy induces human esophageal carcinoma cell pyroptosis by targeting the PKM2/caspase-8/caspase-3/GSDME axis
    Lisha Li, Dongfeng Song, Ling Qi, Mingxia Jiang, Yiming Wu, Junqing Gan, Kui Cao, Yanjing Li, Yuxian Bai, Tongsen Zheng
    Cancer Letters.2021; 520: 143.     CrossRef
  • Retracted: Silence of cZNF292 suppresses the growth, migration, and invasion of human esophageal cancer Eca‐109 cells via upregulating miR‐206
    Zengjia Liu, Guiju Hu, Yan Zhao, Zuorun Xiao, Mingzhe Yan, Mei Ren
    Journal of Cellular Biochemistry.2020; 121(3): 2354.     CrossRef
  • Metformin Promotes Axon Regeneration after Spinal Cord Injury through Inhibiting Oxidative Stress and Stabilizing Microtubule
    Haoli Wang, Zhilong Zheng, Wen Han, Yuan Yuan, Yao Li, Kailiang Zhou, Qingqing Wang, Ling Xie, Ke Xu, Hongyu Zhang, Huazi Xu, Yanqing Wu, Jian Xiao
    Oxidative Medicine and Cellular Longevity.2020; 2020: 1.     CrossRef
  • The caspase-3/GSDME signal pathway as a switch between apoptosis and pyroptosis in cancer
    Mingxia Jiang, Ling Qi, Lisha Li, Yanjing Li
    Cell Death Discovery.2020;[Epub]     CrossRef
  • Metformin delays AKT/c-Met-driven hepatocarcinogenesis by regulating signaling pathways for de novo lipogenesis and ATP generation
    Cong Zhang, Junjie Hu, Lei Sheng, Ming Yuan, Yong Wu, Liang Chen, Guohua Zheng, Zhenpeng Qiu
    Toxicology and Applied Pharmacology.2019; 365: 51.     CrossRef
  • Enhanced penetration and cytotoxicity of metformin and collagenase conjugated gold nanoparticles in breast cancer spheroids
    Elaheh Dalir Abdolahinia, Samad Nadri, Reza Rahbarghazi, Jaleh Barar, Ayoub Aghanejad, Yadollah Omidi
    Life Sciences.2019; 231: 116545.     CrossRef
  • Biguanides Exert Antitumoral Actions in Pituitary Tumor Cells Through AMPK-Dependent and -Independent Mechanisms
    Mari C Vázquez-Borrego, Antonio C Fuentes-Fayos, Aura D Herrera-Martínez, Fernando L-López, Alejandro Ibáñez-Costa, Paloma Moreno-Moreno, María R Alhambra-Expósito, Ana Barrera-Martín, Cristóbal Blanco-Acevedo, Elena Dios, Eva Venegas-Moreno, Juan Soliver
    The Journal of Clinical Endocrinology & Metabolism.2019; 104(8): 3501.     CrossRef
  • Metformin induces TPC-1 cell apoptosis through endoplasmic reticulum stress-associated pathways in vitro and in vivo
    Jianwen Ye, Lei Qi, Kunlun Chen, Renfeng Li, Shengping Song, Chuang Zhou, Wenlong Zhai
    International Journal of Oncology.2019;[Epub]     CrossRef
  • AKT2 contributes to increase ovarian cancer cell migration and invasion through the AKT2-PKM2-STAT3/NF-κB axis
    Bin Zheng, Li Geng, Li Zeng, Fangfang Liu, Qiaojia Huang
    Cellular Signalling.2018; 45: 122.     CrossRef
  • Antitumor effects of metformin are a result of inhibiting nuclear factor kappa B nuclear translocation in esophageal squamous cell carcinoma
    Nobufumi Sekino, Masayuki Kano, Yasunori Matsumoto, Haruhito Sakata, Yasunori Akutsu, Naoyuki Hanari, Kentaro Murakami, Takeshi Toyozumi, Masahiko Takahashi, Ryota Otsuka, Masaya Yokoyama, Tadashi Shiraishi, Koichiro Okada, Isamu Hoshino, Keiko Iida, Aki
    Cancer Science.2018; 109(4): 1066.     CrossRef
  • Down‐regulation of intracellular anti‐apoptotic proteins, particularly c‐FLIP by therapeutic agents; the novel view to overcome resistance to TRAIL
    Ali Hassanzadeh, Majid Farshdousti Hagh, Mohammad Reza Alivand, Ali Akbar Movassaghpour Akbari, Karim Shams Asenjan, Raedeh Saraei, Saeed Solali
    Journal of Cellular Physiology.2018; 233(10): 6470.     CrossRef
  • Interaction Between Prediabetes and the ABO Blood Types in Predicting Postsurgical Esophageal Squamous Cell Carcinoma-Specific Mortality: The FIESTA Study
    Guohui Fan, Dan Hu, Xinran Zhang, Feng Peng, Xiandong Lin, Gang Chen, Binying Liang, Hejun Zhang, Yan Xia, Xiongwei Zheng, Jianzheng Jie, Wenquan Niu
    Frontiers in Oncology.2018;[Epub]     CrossRef
  • Obesity, Diabetes and Gastrointestinal Malignancy: The role of Metformin and other Anti-diabetic Therapy
    McFarlane Samy I
    Global Journal of Obesity, Diabetes and Metabolic Syndrome.2018; : 008.     CrossRef
  • 12,221 View
  • 454 Download
  • 29 Web of Science
  • 26 Crossref
Close layer
Long Non-coding RNA HOXA11 Antisense Promotes Cell Proliferation and Invasion and Predicts Patient Prognosis in Serous Ovarian Cancer
Ga Won Yim, Hee Jung Kim, Lee Kyung Kim, Sang Wun Kim, Sunghoon Kim, Eun Ji Nam, Young Tae Kim
Cancer Res Treat. 2017;49(3):656-668.   Published online October 11, 2016
DOI: https://doi.org/10.4143/crt.2016.263
AbstractAbstract PDFPubReaderePub
Purpose
The biological function of long non-coding RNAs (lncRNAs) is only partially understood; therefore, in this study, we investigated the expression of the novel HOXA11 antisense (HOXA11as) lncRNA and its oncogenic role in serous ovarian cancer (SOC).
Materials and Methods
HOXA11as expression was examined in 129 SOC tissue samples by real time reverse transcription polymerase chain reaction. Clinicopathological factors and patient survival were compared between the high (n=27) and low HOXA11as expression group (n=102). To investigate the role of HOXA11as in cell proliferation, invasion, and migration, HOXA11as expression in ovarian cancer cells was knocked down using RNA interference.
Results
HOXA11as expression in cancer tissue was 77-fold higher than that of noncancerous tissue (p < 0.05). Higher HOXA11as expression was significantly correlated with histological grade (p=0.017) and preoperative cancer antigen 125 (p=0.048). HOXA11as overexpression in SOC cells led to increased cell proliferation, invasion, and migration. Moreover, HOXA11as was associated with the expression of genes involved in cell invasion, migration, and epithelial-mesenchymal transition (EMT), including vascular endothelial growth factor, matrix metalloproteinase 9 (MMP-9), B-catenin, E-cadherin, Snail, Twist, and vimentin. Multivariate analysis revealed that HOXA11as was a prognostic factor of progressive disease and mortality (hazard ratio [HR], 1.730; p=0.043 and HR, 2.170; p=0.033, respectively). Progression-free and overall survival were significantly shorter in patients with high HOXA11as expression.
Conclusion
These findings highlight the clinical significance of HOXA11as to predicting the prognosis of SOC patients and suggest its potential in promoting tumor aggressiveness via regulation of vascular endothelial growth factor (VEGF), MMP-9, and EMT-related mechanisms.

Citations

Citations to this article as recorded by  
  • The Role of EMT-Related lncRNAs in Ovarian Cancer
    Dimitra Ioanna Lampropoulou, Marios Papadimitriou, Christos Papadimitriou, Dimitrios Filippou, Georgia Kourlaba, Gerasimos Aravantinos, Maria Gazouli
    International Journal of Molecular Sciences.2023; 24(12): 10079.     CrossRef
  • LncRNAs and the Angiogenic Switch in Cancer: Clinical Significance and Therapeutic Opportunities
    Peace Mabeta, Rodney Hull, Zodwa Dlamini
    Genes.2022; 13(1): 152.     CrossRef
  • Long non-coding RNA HOXA11-AS knockout inhibits proliferation and overcomes drug resistance in ovarian cancer
    Yuwei Chen, Zhaolei Cui, Qiaoling Wu, Huihui Wang, Hongmei Xia, Yang Sun
    Bioengineered.2022; 13(5): 13893.     CrossRef
  • The role of altered long noncoding RNAs in overall survival of ovarian cancer: A systematic review and meta-analysis
    Elahe Seyed Hosseini, Marziyeh Alizadeh Zarei, Hamed Haddad Kashani, Alireza Milajerdi, Zahra Zare Dehghanani, Hassan Hassani Bafrani, Hossein Nikzad
    Pathology - Research and Practice.2021; 219: 153363.     CrossRef
  • Non-Coding RNAs as Biomarkers of Tumor Progression and Metastatic Spread in Epithelial Ovarian Cancer
    Karolina Seborova, Radka Vaclavikova, Lukas Rob, Pavel Soucek, Pavel Vodicka
    Cancers.2021; 13(8): 1839.     CrossRef
  • Long non-coding RNAs: A view to kill ovarian cancer
    Alexey V. Zamaraev, Pavel I. Volik, Gennady T. Sukhikh, Gelina S. Kopeina, Boris Zhivotovsky
    Biochimica et Biophysica Acta (BBA) - Reviews on Cancer.2021; 1876(1): 188584.     CrossRef
  • Long non-coding RNA CASC15 favors tumorigenesis and development of ovarian cancer via sponging miR-542-3p
    Qiang LI, Wenjing LIU, Sumei LI, Su ZHANG
    Panminerva Medica.2021;[Epub]     CrossRef
  • Long non-coding RNAs in ovarian cancer: expression profile and functional spectrum
    Selin Oncul, Paola Amero, Cristian Rodriguez-Aguayo, George A. Calin, Anil K. Sood, Gabriel Lopez-Berestein
    RNA Biology.2020; 17(11): 1523.     CrossRef
  • A comprehensive review of the role of long non-coding RNAs in organs with an endocrine function
    Soudeh Ghafouri-Fard, Mohammadhosein Esmaeili, Hamed Shoorei, Mohammad Taheri
    Biomedicine & Pharmacotherapy.2020; 125: 110027.     CrossRef
  • Cyclooxygenase-2 promotes ovarian cancer cell migration and cisplatin resistance via regulating epithelial mesenchymal transition
    Lin Deng, Ding-qing Feng, Bin Ling
    Journal of Zhejiang University-SCIENCE B.2020; 21(4): 315.     CrossRef
  • Long Noncoding RNA E2F4as Promotes Progression and Predicts Patient Prognosis in Human Ovarian Cancer
    Sun-Ae Park, Lee Kyung Kim, Young Tae Kim, Tae-Hwe Heo, Hee Jung Kim
    Cancers.2020; 12(12): 3626.     CrossRef
  • lncRNA HOXA11-AS Promotes Proliferation and Migration via Sponging miR-155 in Hypopharyngeal Squamous Cell Carcinoma
    Jianing Xu, Qiyu Bo, Xiang Zhang, Dapeng Lei, Jue Wang, Xinliang Pan
    Oncology Research Featuring Preclinical and Clinical Cancer Therapeutics.2020; 28(3): 311.     CrossRef
  • LncRNAs in ovarian cancer
    Jin-yan Wang, Ai-qing Lu, Li-juan Chen
    Clinica Chimica Acta.2019; 490: 17.     CrossRef
  • Up-regulation of long intergenic noncoding RNA 01296 in ovarian cancer impacts invasion, apoptosis and cell cycle distribution via regulating EMT
    Hui Xu, Jing-Fang Zheng, Cong-Zhe Hou, Yue Li, Pei-Shu Liu
    Cellular Signalling.2019; 62: 109341.     CrossRef
  • A novel risk score system for assessment of ovarian cancer based on co-expression network analysis and expression level of five lncRNAs
    Qian Zhao, Conghong Fan
    BMC Medical Genetics.2019;[Epub]     CrossRef
  • Long non-coding RNA HOTTIP enhances IL-6 expression to potentiate immune escape of ovarian cancer cells by upregulating the expression of PD-L1 in neutrophils
    Anquan Shang, Weiwei Wang, Chenzheng Gu, Chen Chen, Bingjie Zeng, Yibao Yang, Ping Ji, Junjun Sun, Junlu Wu, Wenying Lu, Zujun Sun, Dong Li
    Journal of Experimental & Clinical Cancer Research.2019;[Epub]     CrossRef
  • LINC01210 accelerates proliferation, invasion and migration in ovarian cancer through epigenetically downregulating KLF4
    Chu Zhang, Jie Liu, Yang Zhang, Chengyan Luo, Tong Zhu, Rongrong Zhang, Ruiqin Yao
    Biomedicine & Pharmacotherapy.2019; 119: 109431.     CrossRef
  • Prognostic and clinicopathological significance of long noncoding RNA HOXA11-AS expression in human solid tumors: a meta-analysis
    Shidai Mu, Lisha Ai, Fengjuan Fan, Chunyan Sun, Yu Hu
    Cancer Cell International.2018;[Epub]     CrossRef
  • Long non-coding RNAs in ovarian cancer
    Lei Zhan, Jun Li, Bing Wei
    Journal of Experimental & Clinical Cancer Research.2018;[Epub]     CrossRef
  • HOXA11 antisense long noncoding RNA (HOXA11‐AS): A promising lncRNA in human cancers
    Cheng‐Wei Lu, Dan‐Dan Zhou, Tian Xie, Ji‐Long Hao, Om Prakash Pant, Cheng‐Bo Lu, Xiu‐Fen Liu
    Cancer Medicine.2018; 7(8): 3792.     CrossRef
  • Long noncoding RNA LINC00152 promotes cell proliferation through competitively binding endogenous miR‐125b with MCL‐1 by regulating mitochondrial apoptosis pathways in ovarian cancer
    Puxiang Chen, Xiaolin Fang, Bing Xia, Yan Zhao, Qiaoyan Li, Xiaoying Wu
    Cancer Medicine.2018; 7(9): 4530.     CrossRef
  • Altered long noncoding RNAs and survival outcomes in ovarian cancer
    Li Ning, Ying-chao Hu, Shu Wang, Jing-he Lang
    Medicine.2018; 97(32): e11481.     CrossRef
  • Long non-coding RNAs as emerging regulators of epithelial to mesenchymal transition in gynecologic cancers
    Xiaojing Lin, Junjun Qiu, Keqin Hua
    BioScience Trends.2018; 12(4): 342.     CrossRef
  • RETRACTED ARTICLE: Systematic analyses reveal long non-coding RNA (PTAF)-mediated promotion of EMT and invasion-metastasis in serous ovarian cancer
    Haihai Liang, Xiaoguang Zhao, Chengyu Wang, Jian Sun, Yingzhun Chen, Guoyuan Wang, Lei Fang, Rui Yang, Mengxue Yu, Yunyan Gu, Hongli Shan
    Molecular Cancer.2018;[Epub]     CrossRef
  • LncRNAs KB‑1836B5, LINC00566 and FAM27L are associated with the survival time of patients with ovarian cancer
    Huijian Li, Mi Gong, Min Zhao, Xinru Wang, Wenjun Cheng, Yankai Xia
    Oncology Letters.2018;[Epub]     CrossRef
  • ‘Lnc’‐ing Wnt in female reproductive cancers: therapeutic potential of long non‐coding RNAs in Wnt signalling
    Mei S Ong, Wanpei Cai, Yi Yuan, Hin C Leong, Tuan Z Tan, Asad Mohammad, Ming L You, Frank Arfuso, Boon C Goh, Sudha Warrier, Gautam Sethi, Nicholas S Tolwinski, Peter E Lobie, Celestial T Yap, Shing C Hooi, Ruby Y Huang, Alan P Kumar
    British Journal of Pharmacology.2017; 174(24): 4684.     CrossRef
  • Long non-coding RNA HOXA11-AS in human cancer: A meta-analysis
    Na Li, Meilan Yang, Ke Shi, Wei Li
    Clinica Chimica Acta.2017; 474: 165.     CrossRef
  • Dysregulated expression of homeobox family genes may influence survival outcomes of patients with epithelial ovarian cancer: analysis of data from The Cancer Genome Atlas
    Kyung Jin Eoh, Hee Jung Kim, Jung-Yun Lee, Eun Ji Nam, Sunghoon Kim, Sang Wun Kim, Young Tae Kim
    Oncotarget.2017; 8(41): 70579.     CrossRef
  • 11,672 View
  • 393 Download
  • 35 Web of Science
  • 28 Crossref
Close layer
Effects of Triterpenoid Glycosides from Fresh Ginseng Berry on SW480 Human Colorectal Cancer Cell Line
Jing-Tian Xie, Guang-Jian Du, Eryn McEntee, Han H. Aung, Hui He, Sangeeta R. Mehendale, Chong-Zhi Wang, Chun-Su Yuan
Cancer Res Treat. 2011;43(1):49-55.   Published online March 31, 2011
DOI: https://doi.org/10.4143/crt.2011.43.1.49
AbstractAbstract PDFPubReaderePub
PURPOSE
The pharmacological activities, notably the anticancer properties, of bioactive constituents fromfresh American ginseng berry have not yet been well studied. In this study, we investigated the antiproliferative effects of fresh American ginseng berry extract (AGBE) and its representative triterpenoid glycosides using the human colorectal cancer cell line SW480.
MATERIALS AND METHODS
Using high performance liquid chromatography (HPLC), the contents of 8 ginsenosides in AGBE were determined. The cell growth inhibitory effects of AGBE and three triterpenoid glycosides (ginsenosides Rb3, Re, and Rg3) were evaluated by proliferation assay and 3H-thymidine incorporation assay. Cell cycle and apoptotic effects were analyzed by using flow cytometry after staining with propidium iodide and annexin V.
RESULTS
HPLC analysis data showed that AGBE has a distinct ginsenoside profile. AGBE inhibited SW480 cell growth significantly in a time-dependent (24-96 hours) and concentration-dependent (0.1-1.0 mg/mL) manner. Ginsenosides Rb3, Re, and Rg3 also possess significant antiproliferative activities on SW480 cells. 3H-thymidine incorporation assay indicated that AGBE and ginsenosides Rb3, Re, and Rg3 might inhibit the transferring and duplication of DNA in SW480 cells. Flow cytometric assay data suggested that AGBE arrested SW480 cells in S and G2/M phases, and significantly induced cell apoptosis.
CONCLUSION
AGBE and ginsenosides Rb3, Re, and Rg3 possessed significant antiproliferative effects and induced changes of morphological appearance on SW480 cells. The mechanisms of the antiproliferation of AGBE and tested ginsenosides involved could be cell cycle arrest and induction of apoptosis.

Citations

Citations to this article as recorded by  
  • A systematic review of ginsenoside biosynthesis, spatiotemporal distribution, and response to biotic and abiotic factors in American ginseng
    Lixia Tian, Ranran Gao, Yuxiang Cai, Junxian Chen, Hongmei Dong, Shanshan Chen, Zaichang Yang, Yu Wang, Linfang Huang, Zhichao Xu
    Food & Function.2024; 15(5): 2343.     CrossRef
  • Ginsenoside Re Suppress the Proliferation and Migration of MCF-7 Human Breast Cancer Cells
    Jin-Nyoung Ho
    Journal of the Korean Society of Food Science and Nutrition.2024; 53(3): 233.     CrossRef
  • Comprehensive phytochemicals analysis and anti‐myocardial ischemia activity of total saponins of American ginseng berry
    Le Li, Yunhe Liu, Hui Yu, Zhuo Li, Hongqiang Lin, Fulin Wu, Luying Tan, Caixia Wang, Pingya Li, Jinping Liu
    Journal of Food Biochemistry.2022;[Epub]     CrossRef
  • Pharmacological Properties of Ginsenoside Re
    Xiao-Yan Gao, Guan-Cheng Liu, Jian-Xiu Zhang, Ling-He Wang, Chang Xu, Zi-An Yan, Ao Wang, Yi-Fei Su, Jung-Joon Lee, Guang-Chun Piao, Hai-Dan Yuan
    Frontiers in Pharmacology.2022;[Epub]     CrossRef
  • Therapeutic effects of ginseng and ginsenosides on colorectal cancer
    Linxian Zhao, Yueming Zhang, Yajuan Li, Chen Li, Kai Shi, Kai Zhang, Ning Liu
    Food & Function.2022; 13(12): 6450.     CrossRef
  • Comparative Analysis of Panax ginseng Berries from Seven Cultivars Using UPLC-QTOF/MS and NMR-Based Metabolic Profiling
    Dahye Yoon, Bo-Ram Choi, Young-Chang Kim, Seon Min Oh, Hyoung-Geun Kim, Jang-Uk Kim, Nam-In Baek, Suhkmann Kim, Dae Young Lee
    Biomolecules.2019; 9(9): 424.     CrossRef
  • Multiple Effects of Ginseng Berry Polysaccharides: Plasma Cholesterol Level Reduction and Enteric Neoplasm Prevention
    Jin-Yi Wan, Wei-Hua Huang, Wei Zheng, Chan Woong Park, Su Hwan Kim, Dae Bang Seo, Kwang-Soon Shin, Jinxiang Zeng, Haiqiang Yao, Clara Sava-Segal, Chong-Zhi Wang, Chun-Su Yuan
    The American Journal of Chinese Medicine.2017; 45(06): 1293.     CrossRef
  • Process Optimization of Ginseng Berry Extract Fermentation by Lactobacillus sp. Strain KYH isolated from Fermented Kimchi and Product Analysis
    Yoo-Jin Ha, Sun-Kyun Yoo, Mee Ree Kim
    Journal of the East Asian Society of Dietary Life.2016; 26(1): 88.     CrossRef
  • Anticancer Activities of Protopanaxadiol‐ and Protopanaxatriol‐Type Ginsenosides and Their Metabolites
    Xiao-Jia Chen, Xiao-Jing Zhang, Yan-Mei Shui, Jian-Bo Wan, Jian-Li Gao, Ki-Wan Oh
    Evidence-Based Complementary and Alternative Medicine.2016;[Epub]     CrossRef
  • Metabonomic Profiling Reveals Cancer Chemopreventive Effects of American Ginseng on Colon Carcinogenesis in ApcMin/+ Mice
    Guoxiang Xie, Chong-Zhi Wang, Chunhao Yu, Yunping Qiu, Xiao-Dong Wen, Chun-Feng Zhang, Chun-Su Yuan, Wei Jia
    Journal of Proteome Research.2015; 14(8): 3336.     CrossRef
  • Establishment of Optimal Fermentation Conditions for Steam-dried Ginseng Berry via Friendly Bacteria and Its Antioxidant Activities
    Seung Tae Kim, Hee Jung Kim, Su Kil Jang, Do Ik Lee, Seong Soo Joo
    Korean Journal of Food Science and Technology.2013; 45(1): 77.     CrossRef
  • Ginsenoside F2 induces apoptosis accompanied by protective autophagy in breast cancer stem cells
    Trang Thi Mai, JeongYong Moon, YeonWoo Song, Pham Quoc Viet, Pham Van Phuc, Jung Min Lee, Tae-Hoo Yi, Moonjae Cho, Somi Kim Cho
    Cancer Letters.2012; 321(2): 144.     CrossRef
  • 11,671 View
  • 93 Download
  • 12 Crossref
Close layer
The CXCR4 Antagonist AMD3100 Has Dual Effects on Survival and Proliferation of Myeloma Cells In Vitro
Ha-Yon Kim, Ji-Young Hwang, Seong-Woo Kim, Hyo-Jin Lee, Hwan-Jung Yun, Samyong Kim, Deog-Yeon Jo
Cancer Res Treat. 2010;42(4):225-234.   Published online December 31, 2010
DOI: https://doi.org/10.4143/crt.2010.42.4.225
AbstractAbstract PDFPubReaderePub
Purpose

AMD3100, an antagonist of the CXCR4 chemokine receptor is soon to be used clinically for the peripheral mobilization of hematopoietic stem cells (HSCs) in patients with multiple myeloma. AMD3100 has been shown to activate a G protein coupled with CXCR4 and thus acts as a partial CXCR4 agonist in vitro. Thus, we explored whether AMD3100 affected the survival and proliferation of myeloma cells in vitro.

Materials and Methods

The effects of AMD3100 on survival and proliferation of two myeloma cell lines (RPMI8226 and U266) as well as CD138+ cells obtained from several patients with multiple myeloma were analyzed by flow cytometry using annexin V and a colorimetric cell proliferation assay (CCK-8 assay).

Results

AMD3100, but not T140, another CXCR4 antagonist, stimulated the proliferation of myeloma cell lines and CD138+ primary human myeloma cells (-2-fold increase) in a dose-dependent manner in serum-free culture for up to 5 days, which was inhibited by pretreating the cells with pertussis toxin. AMD3100 enhanced the proliferation of U266 cells induced by interleukin-6 and partially reversed AG490-mediated growth inhibition and apoptosis induced by serum deprivation in RPMI8226 cells. AMD3100 induced the phosphorylation of Akt and MAPK p44/p42 in U266 cells and MAPK p44/p42 in RPMI8226 cells. In contrast, AMD3100 markedly increased the cell apoptosis and reduced the number of RPMI8226 cells after 5 to 7 days of culture under serum-free conditions.

Conclusion

AMD3100 exerts dual effects, initially enhancing and subsequently inhibiting the survival and proliferation of myeloma cells, signaling via CXCR4 in vitro.

Citations

Citations to this article as recorded by  
  • Paradoxical cell targeting of calreticulin-empowered, protein-only nanoparticles
    Eloi Parladé, Annabel García-Leon, Eric Voltà-Durán, Ugutz Unzueta, Ramon Mangues, Isolda Casanova, Antonio Villaverde, Esther Vázquez
    European Journal of Pharmaceutics and Biopharmaceutics.2024; 202: 114410.     CrossRef
  • Lyophilization of biomimetic amyloids preserves their regulatable, endocrine-like functions for nanoparticle release
    Marianna TP Favaro, Hèctor López-Laguna, Eric Voltà-Durán, Lorena Alba-Castellon, Julieta M. Sánchez, Isolda Casanova, Ugutz Unzueta, Ramón Mangues, Antonio Villaverde, Esther Vázquez
    Applied Materials Today.2024; 39: 102348.     CrossRef
  • Time-Prolonged Release of Tumor-Targeted Protein–MMAE Nanoconjugates from Implantable Hybrid Materials
    Naroa Serna, Aïda Falgàs, Annabel García-León, Ugutz Unzueta, Yáiza Núñez, Alejandro Sánchez-Chardi, Carlos Martínez-Torró, Ramón Mangues, Esther Vazquez, Isolda Casanova, Antonio Villaverde
    Pharmaceutics.2022; 14(1): 192.     CrossRef
  • Non-invasive detection and complementary diagnosis of liver metastases via chemokine receptor 4 imaging
    Hua Yang, Shanshan Tan, Jingjuan Qiao, Yiting Xu, Zongxiang Gui, Yuguang Meng, Bin Dong, Guangda Peng, Oluwatosin Y. Ibhagui, Weiping Qian, Jimmy Lu, Zezhong Li, Guimin Wang, Jinping Lai, Lily Yang, Hans E. Grossniklaus, Jenny J. Yang
    Cancer Gene Therapy.2022; 29(12): 1827.     CrossRef
  • Controlling self-assembling and tumor cell-targeting of protein-only nanoparticles through modular protein engineering
    Eric Voltà-Durán, Olivia Cano-Garrido, Naroa Serna, Hèctor López-Laguna, Laura Sánchez-García, Mireia Pesarrodona, Alejandro Sánchez-Chardi, Ramón Mangues, Antonio Villaverde, Esther Vázquez, Ugutz Unzueta
    Science China Materials.2020; 63(1): 147.     CrossRef
  • Engineering Secretory Amyloids for Remote and Highly Selective Destruction of Metastatic Foci
    María Virtudes Céspedes, Olivia Cano‐Garrido, Patricia Álamo, Rita Sala, Alberto Gallardo, Naroa Serna, Aïda Falgàs, Eric Voltà‐Durán, Isolda Casanova, Alejandro Sánchez‐Chardi, Hèctor López‐Laguna, Laura Sánchez‐García, Julieta M. Sánchez, Ugutz Unzueta,
    Advanced Materials.2020;[Epub]     CrossRef
  • Recruiting potent membrane penetrability in tumor cell-targeted protein-only nanoparticles
    Naroa Serna, Julieta M Sánchez, Ugutz Unzueta, Laura Sánchez-García, Alejandro Sánchez-Chardi, Ramón Mangues, Esther Vázquez, Antonio Villaverde
    Nanotechnology.2019; 30(11): 115101.     CrossRef
  • The Involvement of SDF-1α/CXCR4 Axis in Radiation-Induced Acute Injury and Fibrosis of Skin
    Jinming Cao, Wei Zhu, Daojiang Yu, Lu Pan, Li Zhong, Yuji Xiao, Yiying Gao, Yang Jiao, Qi Zhang, Jiang Ji, Hongying Yang, Shuyu Zhang, Jianping Cao
    Radiation Research.2019; 192(4): 410.     CrossRef
  • Chemokine-mimetic plerixafor derivative for tumor-specific delivery of nanomaterials
    Seungbeom Ko, Gayong Shim, Jinyoung Kim, Yu-Kyoung Oh
    Nano Research.2018; 11(4): 2159.     CrossRef
  • Protein Nanoparticles are nontoxic, Tuneable Cell Stressors
    Marianna Teixeira de Pinho Favaro, Laura Sánchez-García, Alejandro Sánchez-Chardi, Mónica Roldán, Ugutz Unzueta, Naroa Serna, Olivia Cano-Garrido, Adriano Rodrigues Azzoni, Neus Ferrer-Miralles, Antonio Villaverde, Esther Vázquez
    Nanomedicine.2018; 13(3): 255.     CrossRef
  • Self-assembling toxin-based nanoparticles as self-delivered antitumoral drugs
    Laura Sánchez-García, Naroa Serna, Patricia Álamo, Rita Sala, María Virtudes Céspedes, Mònica Roldan, Alejandro Sánchez-Chardi, Ugutz Unzueta, Isolda Casanova, Ramón Mangues, Esther Vázquez, Antonio Villaverde
    Journal of Controlled Release.2018; 274: 81.     CrossRef
  • The CXCR4 antagonist plerixafor (AMD3100) promotes proliferation of Ewing sarcoma cell lines in vitro and activates receptor tyrosine kinase signaling
    Philipp Berning, Christiane Schaefer, Dagmar Clemens, Eberhard Korsching, Uta Dirksen, Jenny Potratz
    Cell Communication and Signaling.2018;[Epub]     CrossRef
  • Switching cell penetrating and CXCR4-binding activities of nanoscale-organized arginine-rich peptides
    Marianna Teixeira de Pinho Favaro, Naroa Serna, Laura Sánchez-García, Rafael Cubarsi, Mónica Roldán, Alejandro Sánchez-Chardi, Ugutz Unzueta, Ramón Mangues, Neus Ferrer-Miralles, Adriano Rodrigues Azzoni, Esther Vázquez, Antonio Villaverde
    Nanomedicine: Nanotechnology, Biology and Medicine.2018; 14(6): 1777.     CrossRef
  • The inhibitory effects and mechanisms of 3,6-O-sulfated chitosan against human papillomavirus infection
    Yanyun Gao, Wei Liu, Wei Wang, Xiaoshuang Zhang, Xia Zhao
    Carbohydrate Polymers.2018; 198: 329.     CrossRef
  • CXCL12/CXCR4/CXCR7 Chemokine Axis in the Central Nervous System: Therapeutic Targets for Remyelination in Demyelinating Diseases
    Tianci Chu, Lisa B. E. Shields, Yi Ping Zhang, Shi-Qing Feng, Christopher B. Shields, Jun Cai
    The Neuroscientist.2017; 23(6): 627.     CrossRef
  • Hypoxia and hyperoxia differentially control proliferation of rat neural crest stem cells via distinct regulatory pathways of the HIF1α–CXCR4 and TP53–TPM1 proteins
    Chien‐Cheng Chen, Ching‐Wu Hsia, Cheng‐Wen Ho, Chang‐Min Liang, Chieh‐Min Chen, Kun‐Lun Huang, Bor‐Hwang Kang, Yi‐Hui Chen
    Developmental Dynamics.2017; 246(3): 162.     CrossRef
  • CXCL12 and CXCR7 are relevant targets to reverse cell adhesion‐mediated drug resistance in multiple myeloma
    Johannes M. Waldschmidt, Anna Simon, Dagmar Wider, Stefan J. Müller, Marie Follo, Gabriele Ihorst, Sarah Decker, Joschka Lorenz, Manik Chatterjee, Abdel K. Azab, Justus Duyster, Ralph Wäsch, Monika Engelhardt
    British Journal of Haematology.2017; 179(1): 36.     CrossRef
  • CXCR4 blockade with AMD3100 enhances Taxol chemotherapy to limit ovarian cancer cell growth
    Patrick M. Reeves, Mojgan A. Abbaslou, Farah R.W. Kools, Mark C. Poznansky
    Anti-Cancer Drugs.2017; 28(9): 935.     CrossRef
  • AMD3100: A Versatile Platform for CXCR4 Targeting 68Ga-Based Radiopharmaceuticals
    Sophie Poty, Eleni Gourni, Pauline Désogère, Frédéric Boschetti, Christine Goze, Helmut R. Maecke, Franck Denat
    Bioconjugate Chemistry.2016; 27(3): 752.     CrossRef
  • The CXCR4 antagonist plerixafor enhances the effect of rituximab in diffuse large B-cell lymphoma cell lines
    Linn Reinholdt, Maria Bach Laursen, Alexander Schmitz, Julie Støve Bødker, Lasse Hjort Jakobsen, Martin Bøgsted, Hans Erik Johnsen, Karen Dybkær
    Biomarker Research.2016;[Epub]     CrossRef
  • PCAF-mediated Akt1 acetylation enhances the proliferation of human glioblastoma cells
    Shuguang Zhang, Guan Sun, Zhimin Wang, Yi Wan, Jun Guo, Lei Shi
    Tumor Biology.2015; 36(3): 1455.     CrossRef
  • Chemokines and Bone
    Annette Gilchrist, Paula H. Stern
    Clinical Reviews in Bone and Mineral Metabolism.2015; 13(2): 61.     CrossRef
  • AMD3100 reduces CXCR4-mediated survival and metastasis of osteosarcoma by inhibiting JNK and Akt, but not p38 or Erk1/2, pathways in in vitro and mouse experiments
    YU-XIN LIAO, ZE-ZE FU, CHENG-HAO ZHOU, LIAN-CHENG SHAN, ZHUO-YING WANG, FEI YIN, LONG-PO ZHENG, YING-QI HUA, ZHENG-DONG CAI
    Oncology Reports.2015; 34(1): 33.     CrossRef
  • CXC Chemokine Ligand 12 Protects Pancreatic β-Cells from Necrosis through Akt Kinase-Mediated Modulation of Poly(ADP-ribose) Polymerase-1 Activity
    Nevena Grdović, Svetlana Dinić, Mirjana Mihailović, Aleksandra Uskoković, Jelena Arambašić Jovanović, Goran Poznanović, Ludwig Wagner, Melita Vidaković, Malka Cohen-Armon
    PLoS ONE.2014; 9(7): e101172.     CrossRef
  • Potential therapeutic biomarkers in plasma cell myeloma: A flow cytometry study
    Wenli Zheng, Dingshen Liu, Xiangshan Fan, Linda Powers, Maitrayee Goswami, Ying Hu, Pei Lin, L. Jeffrey Medeiros, Sa A. Wang
    Cytometry Part B: Clinical Cytometry.2013; 84B(4): 222.     CrossRef
  • Targeting Paraprotein Biosynthesis for Non-Invasive Characterization of Myeloma Biology
    Katharina Lückerath, Constantin Lapa, Annika Spahmann, Gerhard Jörg, Samuel Samnick, Andreas Rosenwald, Herrmann Einsele, Stefan Knop, Andreas K. Buck, Evren Alici
    PLoS ONE.2013; 8(12): e84840.     CrossRef
  • An Antedrug of the CXCL12 Neutraligand Blocks Experimental Allergic Asthma without Systemic Effect in Mice
    François Daubeuf, Muriel Hachet-Haas, Patrick Gizzi, Vincent Gasparik, Dominique Bonnet, Valérie Utard, Marcel Hibert, Nelly Frossard, Jean-Luc Galzi
    Journal of Biological Chemistry.2013; 288(17): 11865.     CrossRef
  • EETs and CYP2J2 inhibit TNF-α-induced apoptosis in pulmonary artery endothelial cells and TGF-β1-induced migration in pulmonary artery smooth muscle cells
    WENJING FENG, XIZHEN XU, GANG ZHAO, GENG LI, TIANTIAN LIU, JUNJIE ZHAO, RUOLAN DONG, DAO WEN WANG, LING TU
    International Journal of Molecular Medicine.2013; 32(3): 685.     CrossRef
  • The role of the CXCL12-CXCR4/CXCR7 axis in the progression and metastasis of bone sarcomas (Review)
    YU-XIN LIAO, CHENG-HAO ZHOU, HUI ZENG, DONG-QING ZUO, ZHUO-YING WANG, FEI YIN, YING-QING HUA, ZHENG-DONG CAI
    International Journal of Molecular Medicine.2013; 32(6): 1239.     CrossRef
  • Why and how to find neutraligands targeting chemokines?
    Jean-Luc Galzi, Muriel Haas, Nelly Frossard, Marcel Hibert
    Drug Discovery Today: Technologies.2012; 9(4): e245.     CrossRef
  • Sublytic C5b‐9 complexes induce proliferative changes of glomerular mesangial cells in rat Thy‐1 nephritis through TRAF6‐mediated PI3K‐dependent Akt1 activation
    Wen Qiu, Yan Zhang, Xiaomei Liu, Jianbo Zhou, Yan Li, Ying Zhou, Kai Shan, Mei Xia, Nan Che, Xuefeng Feng, Dan Zhao, Yingwei Wang
    The Journal of Pathology.2012; 226(4): 619.     CrossRef
  • Initial Assessment of the Role of CXC Chemokine Receptor 4 after Polytrauma
    Harold H. Bach, Vikas Saini, Todd A. Baker, Abhishek Tripathi, Richard L. Gamelli, Matthias Majetschak
    Molecular Medicine.2012; 18(7): 1056.     CrossRef
  • Differential effects of CXCR4 antagonists on the survival and proliferation of myeloid leukemia cellsin vitro
    Ha-Yon Kim, Ji-Young Hwang, Yoon-Suk Oh, Seong-Woo Kim, Hyo-Jin Lee, Hwan-Jung Yun, Samyong Kim, Young-Jun Yang, Deog-Yeon Jo
    The Korean Journal of Hematology.2011; 46(4): 244.     CrossRef
  • 11,709 View
  • 80 Download
  • 33 Crossref
Close layer
Review Article
HIF-1α: a Valid Therapeutic Target for Tumor Therapy
Soon-Sun Hong, Hyunseung Lee, Kyu-Won Kim
Cancer Res Treat. 2004;36(6):343-353.   Published online December 31, 2004
DOI: https://doi.org/10.4143/crt.2004.36.6.343
AbstractAbstract PDFPubReaderePub

Hypoxia plays a major role in the induction of angiogenesis during tumor development. One mechanism by which tumor cells respond to a reduced oxygen level is via the activation of hypoxia-inducible factor-1 (HIF-1). HIF-1 is an oxygen-dependent transcriptional activator that plays crucial roles in the angiogenesis of tumors and mammalian development. HIF-1 consists of a constitutively expressed HIF-1β subunit and the highly regulated HIF-1α subunits. The stability and activity of HIF-1α are regulated by various post-translational modifications, hydroxylation, acetylation, phosphorylation and sumoyaltion. Therefore, HIF-1α interacts with several protein factors including PHD, pVHL, ARD-1, SUMO and p300/CBP. Under normoxia, the HIF-1α subunit is rapidly degraded via the von Hippel-Lindau tumor suppressor gene product (pVHL)-mediated ubiquitin/proteasome pathway. The association of pVHL and HIF-1α under normoxic conditions is triggered by the hydroxylation of prolines and the acetylation of lysine within a polypeptide segment known as the oxygen-dependent degradation (ODD) domain. On the contrary, under the hypoxia condition, the HIF-1α subunit becomes stable and interacts with coactivators such as p300/CBP to modulate its transcriptional activity. Under hypoxic conditions, HIF-1 eventually acts as a master regulator of numerous hypoxia-inducible genes. The target genes of HIF-1 are especially related to angiogenesis, cell proliferation and survival, and to glucose and iron metabolism. Moreover, it was reported that the activation of HIF-1α is closely associated with a variety of tumors and oncogenic pathways. Hence, the blocking of HIF-1α itself or the blocking of HIF-1α interacting proteins inhibits tumor growth. Based on these findings, HIF-1 can be a prime target for anticancer therapies. Therefore, this review summarizes the molecular mechanism of HIF-1α stability, the biological functions of HIF-1 and its potential applications for cancer therapies.

Citations

Citations to this article as recorded by  
  • A comprehensive review on therapeutic potentials of photobiomodulation for neurodegenerative disorders
    Pooja Ramakrishnan, Aradhana Joshi, Mohamed Fazil, Pankaj Yadav
    Life Sciences.2024; 336: 122334.     CrossRef
  • Novel Benzochromenes: Design, Synthesis, Cytotoxicity, Molecular Docking and Mechanistic Investigations
    Fatma G Abdulrahman, Hamada S Abulkhair, Riham A Zidan, Asmaa I Alwakeel, Ahmed A Al-Karmalawy, Ebtehal M Husseiny
    Future Medicinal Chemistry.2024; 16(2): 105.     CrossRef
  • Abnormal changes in metabolites caused by m6A methylation modification: The leading factors that induce the formation of immunosuppressive tumor microenvironment and their promising potential for clinical application
    Liang Zhao, Junchen Guo, Shasha Xu, Meiqi Duan, Baiming Liu, He Zhao, Yihan Wang, Haiyang liu, Zhi Yang, Hexue Yuan, Xiaodi Jiang, Xiaofeng Jiang
    Journal of Advanced Research.2024;[Epub]     CrossRef
  • Role of natural secondary metabolites as HIF-1 inhibitors in cancer therapy
    Prem Shankar Mishra, Rakhi Mishra, Vaishali Manikrao Patil, Samiksha Dewangan
    Medicinal Chemistry Research.2024; 33(5): 721.     CrossRef
  • SIMULATED AEROMEDICAL EVACUATION EXACERBATES ACUTE LUNG INJURY VIA HYPOXIA-INDUCIBLE FACTOR-1α-MEDIATED BNIP3/NIX-DEPENDENT MITOPHAGY
    Chunli Yin, Xinyang Liu, Yujie Ma, Zibo Tang, Wenmin Guo, Bingbing Sun, Jingmei He
    Shock.2024;[Epub]     CrossRef
  • Inferring upstream regulatory genes of FOXP3 in human regulatory T cells from time-series transcriptomic data
    Stefano Magni, Rucha Sawlekar, Christophe M. Capelle, Vera Tslaf, Alexandre Baron, Ni Zeng, Laurent Mombaerts, Zuogong Yue, Ye Yuan, Feng Q. Hefeng, Jorge Gonçalves
    npj Systems Biology and Applications.2024;[Epub]     CrossRef
  • Stilbenoid derivatives as potent inhibitors of HIF-1α-centric cancer metabolism under hypoxia
    Tae-Hee Han, Joohan Lee, Dipesh S. Harmalkar, Hyeseul Kang, Guanghai Jin, Min Kyung Park, Minkyoung Kim, Hyun-A Yang, Jinsu Kim, Su Jeong Kwon, Tae-Su Han, Yongseok Choi, Misun Won, Hyun Seung Ban, Kyeong Lee
    Biomedicine & Pharmacotherapy.2024; 176: 116838.     CrossRef
  • Targeting Hypoxia and HIF1α in Triple-Negative Breast Cancer: New Insights from Gene Expression Profiling and Implications for Therapy
    Delong Han, Zeyu Li, Lingjie Luo, Hezhong Jiang
    Biology.2024; 13(8): 577.     CrossRef
  • The Effects of Normobaric Hypoxia on the Acute Physiological Responses to Resistance Training: A Narrative Review
    Giselle L. Allsopp, Florian A. Britto, Craig R. Wright, Louise Deldicque
    Journal of Strength & Conditioning Research.2024;[Epub]     CrossRef
  • Physiological hypoxia promotes cancer cell migration and attenuates angiogenesis in co-culture using a microfluidic device
    Satoshi Aratake, Kenichi Funamoto
    Microfluidics and Nanofluidics.2024;[Epub]     CrossRef
  • Design, synthesis, molecular dynamics and gene silencing studies of novel therapeutic HIF-1α siRNAs in hypoxic cancer cells
    Jyoti Singh, Sneha Yadav, Archana Bharti Sonkar, Anurag Kumar, Neeraj Kumar Shrivastava, Rohit Kumar, Dharmendra Kumar, Mohd Nazam Ansari, Abdulaziz S. Saeedan, Gaurav Kaithwas
    International Journal of Biological Macromolecules.2024; 282: 136943.     CrossRef
  • Concise Asymmetric Total Syntheses of (+)-Dihydropleurotinic Acid and (−)-Pleurotin, Enabling Rapid Late-Stage Diversification
    Bin Huang, Jing Pang, Nan Cao, Ya-Shuang Dai, Ya-Qiu Long
    JACS Au.2024; 4(11): 4206.     CrossRef
  • Targeting HIF-1 for prostate cancer: a synthesis of preclinical evidence
    Yarden Zohar, Nicola J. Mabjeesh
    Expert Opinion on Therapeutic Targets.2023; 27(8): 715.     CrossRef
  • Transcriptome Analysis on Hepatopancreas Reveals the Metabolic Dysregulation Caused by Vibrio parahaemolyticus Infection in Litopenaeus vannamei
    Miao Miao, Shihao Li, Yuan Liu, Yang Yu, Fuhua Li
    Biology.2023; 12(3): 417.     CrossRef
  • Determination of HIF-1α degradation pathways via modulation of the propionyl mark
    Kwanyoung Jeong, Jinmi Choi, Ahrum Choi, Joohee Shim, Young Ah Kim, Changseok Oh, Hong-Duk Youn, Eun-Jung Cho
    BMB Reports.2023; 56(4): 252.     CrossRef
  • Baf155 regulates skeletal muscle metabolism via HIF-1a signaling
    Jong-Seol Kang, Dongha Kim, Joonwoo Rhee, Ji-Yun Seo, Inkuk Park, Ji-Hoon Kim, Daewon Lee, WonUk Lee, Ye Lynne Kim, Kyusang Yoo, Sunghwan Bae, Jongkyeong Chung, Rho Hyun Seong, Young-Yun Kong, Simon M. Hughes
    PLOS Biology.2023; 21(7): e3002192.     CrossRef
  • Genetic and pharmaceutical targeting of HIF1α allows combo-immunotherapy to boost graft vs. leukemia without exacerbation graft vs. host disease
    Christopher Bailey, Yuanyi Wei, Jinsong Yan, Dan Huang, Peng Zhang, Chong Qi, Christopher Lazarski, JuanJuan Su, Fei Tang, Chun-shu Wong, Pan Zheng, Yan Liu, Yang Liu, Yin Wang
    Cell Reports Medicine.2023; 4(11): 101236.     CrossRef
  • Assessment of hypoxia and oxidative-related changes in a lung-derived brain metastasis model by [64Cu][Cu(ATSM)] PET and proteomic studies
    Jade Fantin, Jérôme Toutain, Elodie A. Pérès, Benoit Bernay, Sarina Maya Mehani, Charly Helaine, Mickael Bourgeois, Carole Brunaud, Laurent Chazalviel, Julien Pontin, Aurélien Corroyer-Dulmont, Samuel Valable, Michel Cherel, Myriam Bernaudin
    EJNMMI Research.2023;[Epub]     CrossRef
  • Exploring mRNA Translation Strategies for Hypoxia Adaptation Across Distantly Related Metazoans
    Manfei Liang, Qingyou Zhao, Yixiang Song, Jianchao Chen
    Epigenomics.2023; 15(20): 1069.     CrossRef
  • Novel thioether linked 4-hydroxycoumarin derivatives: Synthesis, characterization, in vitro pharmacological investigation and molecular docking studies
    B. Manjunatha, Yadav D. Bodke, H.M. Kumaraswamy, P. Meghana, O. Nagaraja, G.C. Anjan kumar
    Journal of Molecular Structure.2022; 1249: 131642.     CrossRef
  • A Concise Synthesis of Pleurotin Enabled by a Nontraditional C–H Epimerization
    John F. Hoskin, Erik J. Sorensen
    Journal of the American Chemical Society.2022; 144(31): 14042.     CrossRef
  • Regulatory mechanisms and function of hypoxia-induced long noncoding RNA NDRG1-OT1 in breast cancer cells
    Hsing-Hua Chao, Jun-Liang Luo, Ming-Hsuan Hsu, Li-Han Chen, Tzu-Pin Lu, Mong-Hsun Tsai, Eric Y. Chuang, Li-Ling Chuang, Liang-Chuan Lai
    Cell Death & Disease.2022;[Epub]     CrossRef
  • A dynamic fluorescence probed glycolysis suppression process in HeLa cells treated with trichostatin A
    Fengqiu Zhang, Changsheng Shao, Yahui Wu, Wei Zhao, Xumiao Jing, Cao Fang, Qing Huang
    The Analyst.2022; 147(22): 5082.     CrossRef
  • Polystyrene-Based Slippery Surfaces Enable the Generation and Easy Retrieval of Tumor Spheroids
    Priyanka Pulugu, Neha Arya, Prasoon Kumar, Akshay Srivastava
    ACS Applied Bio Materials.2022; 5(12): 5582.     CrossRef
  • Prognostic Significance of VEGF and HIF-1 α in Hepatocellular Carcinoma Patients Receiving Sorafenib Versus Metformin Sorafenib Combination
    Shereen El Shorbagy, Fouad abuTaleb, Hany A. Labib, Huda Ebian, Ola A. Harb, Mona Saeed Mohammed, Hanaa A. Rashied, Khaled A. Elbana, Rasha Haggag
    Journal of Gastrointestinal Cancer.2021; 52(1): 269.     CrossRef
  • Adenylate Kinase 4 Promotes Inflammatory Gene Expression via Hif1α and AMPK in Macrophages
    Wei-Yao Chin, Chi-Ying He, Tsun Wai Chow, Qi-You Yu, Liang-Chuan Lai, Shi-Chuen Miaw
    Frontiers in Immunology.2021;[Epub]     CrossRef
  • Stabilization of Hypoxia-Inducible Factor Promotes Antimicrobial Activity of Human Macrophages Against Mycobacterium tuberculosis
    Sebastian F. Zenk, Sebastian Hauck, Daniel Mayer, Mark Grieshober, Steffen Stenger
    Frontiers in Immunology.2021;[Epub]     CrossRef
  • BICD Cargo Adaptor 1 (BICD1) Downregulation Correlates with a Decreased Level of PD-L1 and Predicts a Favorable Prognosis in Patients with IDH1-Mutant Lower-Grade Gliomas
    Shang-Pen Huang, Chien-Hsiu Li, Wei-Min Chang, Yuan-Feng Lin
    Biology.2021; 10(8): 701.     CrossRef
  • The cytotoxic effect of Baeckea frustescens extracts in eliminating hypoxic breast cancer cells
    S. H. Shahruzaman, F. Z. Yusof, S. Maniam, S. Fakurazi, S. Maniam
    BMC Complementary Medicine and Therapies.2021;[Epub]     CrossRef
  • A highly annotated database of genes associated with platinum resistance in cancer
    Dongqing Huang, Sara R. Savage, Anna P. Calinawan, Chenwei Lin, Bing Zhang, Pei Wang, Timothy K. Starr, Michael J. Birrer, Amanda G. Paulovich
    Oncogene.2021; 40(46): 6395.     CrossRef
  • Lung Adenocarcinoma Transcriptomic Analysis Predicts Adenylate Kinase Signatures Contributing to Tumor Progression and Negative Patient Prognosis
    Jonathan A. Chacon-Barahona, Ivan A. Salladay-Perez, Nathan James Lanning
    Metabolites.2021; 11(12): 859.     CrossRef
  • Clinicopathological significance and angiogenic role of the constitutive phosphorylation of the FOXO1 transcription factor in colorectal cancer
    Young San Ko, Nae Yu Kim, Jung-Soo Pyo
    Pathology - Research and Practice.2020; 216(11): 153150.     CrossRef
  • Role of succinate dehydrogenase deficiency and oncometabolites in gastrointestinal stromal tumors
    Yue Zhao, Fei Feng, Qing-Hong Guo, Yu-Ping Wang, Rui Zhao
    World Journal of Gastroenterology.2020; 26(34): 5074.     CrossRef
  • Still Living Better through Chemistry: An Update on Caloric Restriction and Caloric Restriction Mimetics as Tools to Promote Health and Lifespan
    Carla Almendáriz-Palacios, Darrell D. Mousseau, Christopher H. Eskiw, Zoe E. Gillespie
    International Journal of Molecular Sciences.2020; 21(23): 9220.     CrossRef
  • Long Noncoding RNA Expression Profiling Reveals Upregulation of Uroplakin 1A and Uroplakin 1A Antisense RNA 1 under Hypoxic Conditions in Lung Cancer Cells
    Yuree Byun, Young-Chul Choi, Yongsu Jeong, Jaeseung Yoon, Kwanghee Baek
    Molecules and Cells.2020; 43(12): 975.     CrossRef
  • Convergent evolution on the hypoxia-inducible factor (HIF) pathway genes EGLN1 and EPAS1 in high-altitude ducks
    Allie M. Graham, Kevin G. McCracken
    Heredity.2019; 122(6): 819.     CrossRef
  • Inhibitory effects of plasmid small interfering RNA targeting signal transducer and activator of transcription‑3 in C6 glioma cells
    Jiajun Chen, Yue Zhou, Xiangyu Zhou, Jia Li, Feng Liu, Qingwei Zhou
    Molecular Medicine Reports.2019;[Epub]     CrossRef
  • Therapeutic Angiogenesis of Chinese Herbal Medicines in Ischemic Heart Disease: A Review
    Dongqing Guo, Colin E. Murdoch, Tianhua Liu, Jia Qu, Shihong Jiao, Yong Wang, Wei Wang, Xing Chen
    Frontiers in Pharmacology.2018;[Epub]     CrossRef
  • ARD1/NAA10 in hepatocellular carcinoma: pathways and clinical implications
    Danbi Lee, Myoung-Kuk Jang, Ji Hae Seo, Soo Hyung Ryu, Jeong A. Kim, Young-Hwa Chung
    Experimental & Molecular Medicine.2018; 50(7): 1.     CrossRef
  • Low dose radiation primed iNOS + M1macrophages modulate angiogenic programming of tumor derived endothelium
    Vinod Nadella, Sandhya Singh, Aklank Jain, Manju Jain, Karen M. Vasquez, Ashok Sharma, Pranay Tanwar, Goura Kishore Rath, Hridayesh Prakash
    Molecular Carcinogenesis.2018; 57(11): 1664.     CrossRef
  • HIF-1 as a Marker of Age-Related Diseases Associated with Tissue Hypoxia
    E. S. Popravka, N. S. Linkova, S. V. Trofimova, V. Kh. Khavinson
    Biology Bulletin Reviews.2018; 8(6): 497.     CrossRef
  • HRAS as a potential therapeutic target of salirasib RAS inhibitor in bladder cancer
    Satoshi Sugita, Hideki Enokida, Hirofumi Yoshino, Kazutaka Miyamoto, Masaya Yonemori, Takashi Sakaguchi, Yoichi Osako, Masayuki Nakagawa
    International Journal of Oncology.2018;[Epub]     CrossRef
  • HIF-1α contributes to tube malformation of human lymphatic endothelial cells by upregulating VEGFR-3
    Tao Han, Jun Yan, Haini Chen, Yi Ji, Jianbing Chen, Jie Cui, Weimin Shen, Jijun Zou
    International Journal of Oncology.2018;[Epub]     CrossRef
  • Silencing Ubc9 expression suppresses osteosarcoma tumorigenesis and enhances chemosensitivity to HSV-TK/GCV by regulating connexin�43 SUMOylation
    Dianying Zhang, Kai Yu, Zhong Yang, Yanxia Li, Xiaofang Ma, Xiyun Bian, Fengting Liu, Lili Li, Xiaozhi Liu, Wenhan Wu
    International Journal of Oncology.2018;[Epub]     CrossRef
  • BP-1T, an antiangiogenic benzophenone-thiazole pharmacophore, counteracts HIF-1 signalling through p53/MDM2-mediated HIF-1α proteasomal degradation
    Prabhu Thirusangu, V. Vigneshwaran, T. Prashanth, B. R. Vijay Avin, Vikas H. Malojirao, H. Rakesh, Shaukath Ara Khanum, Riaz Mahmood, B. T. Prabhakar
    Angiogenesis.2017; 20(1): 55.     CrossRef
  • Hypoxia inducible factor down-regulation, cancer and cancer stem cells (CSCs): ongoing success stories
    Anthony R. Martin, Cyril Ronco, Luc Demange, Rachid Benhida
    MedChemComm.2017; 8(1): 21.     CrossRef
  • A tumoural angiogenic gateway blocker, Benzophenone-1B represses the HIF-1α nuclear translocation and its target gene activation against neoplastic progression
    Prabhu Thirusangu, V. Vigneshwaran, V. Lakshmi Ranganatha, B.R. Vijay Avin, Shaukath Ara Khanum, Riaz Mahmood, K. Jayashree, B.T. Prabhakar
    Biochemical Pharmacology.2017; 125: 26.     CrossRef
  • Shikonin suppresses proliferation and induces cell cycle arrest through the inhibition of hypoxia-inducible factor-1α signaling
    Ming Yue Li, Chunliu Mi, Ke Si Wang, Zhe Wang, Hong Xiang Zuo, Lian Xun Piao, Guang Hua Xu, Xuezheng Li, Juan Ma, Xuejun Jin
    Chemico-Biological Interactions.2017; 274: 58.     CrossRef
  • Nuclear translocation of HIF-1α induced by influenza A (H1N1) infection is critical to the production of proinflammatory cytokines
    Xinkun Guo, Zhaoqin Zhu, Wanju Zhang, Xiaoxiao Meng, Yong Zhu, Peng Han, Xiaohui Zhou, Yunwen Hu, Ruilan Wang
    Emerging Microbes & Infections.2017; 6(1): 1.     CrossRef
  • Hypoxia-inducible factor-1α in Antarctic notothenioids contains a polyglutamine and glutamic acid insert that varies in length with phylogeny
    A. S. Rix, T. J. Grove, K. M. O’Brien
    Polar Biology.2017; 40(12): 2537.     CrossRef
  • 125I seed irradiation induces apoptosis and inhibits angiogenesis by decreasing HIF-1α and VEGF expression in lung carcinoma xenografts
    Gui-Ling Xiang, Xin-Hong Zhu, Cun-Zhi Lin, Li-Jun Wang, Yong Sun, Yi-Wei Cao, Fang-Fang Wang
    Oncology Reports.2017; 37(5): 3075.     CrossRef
  • Forkhead Transcription Factor FOXO1 Inhibits Angiogenesis in Gastric Cancer in Relation to SIRT1
    Sue Youn Kim, Young San Ko, Jinju Park, Yiseul Choi, Jong-Wan Park, Younghoon Kim, Jung-Soo Pyo, Young Bok Yoo, Jae-Seon Lee, Byung Lan Lee
    Cancer Research and Treatment.2016; 48(1): 345.     CrossRef
  • Aryl Hydrocarbon Receptor Activates NDRG1 Transcription under Hypoxia in Breast Cancer Cells
    En-Yu Li, Wei-Yung Huang, Ya-Chu Chang, Mong-Hsun Tsai, Eric Y. Chuang, Qian-Yu Kuok, Shih-Ting Bai, Lo-Yun Chao, Yuh-Pyng Sher, Liang-Chuan Lai
    Scientific Reports.2016;[Epub]     CrossRef
  • Transcriptional regulation of human and murine short-chain dehydrogenase/reductases (SDRs) – an in silico approach
    Bettina Ebert, Michael Kisiela, Edmund Maser
    Drug Metabolism Reviews.2016; 48(2): 183.     CrossRef
  • Arsenic trioxide plus PX-478 achieves effective treatment in pancreatic ductal adenocarcinoma
    Mingxiao Lang, Xiuchao Wang, Hongwei Wang, Jie Dong, Chungen Lan, Jihui Hao, Chongbiao Huang, Xin Li, Ming Yu, Yanhui Yang, Shengyu Yang, He Ren
    Cancer Letters.2016; 378(2): 87.     CrossRef
  • Hypoxia Positively Regulates the Expression of pH-Sensing G-Protein–Coupled Receptor OGR1 (GPR68)
    Cheryl de Vallière, Jesus Cosin-Roger, Simona Simmen, Kirstin Atrott, Hassan Melhem, Jonas Zeitz, Mehdi Madanchi, Irina Tcymbarevich, Michael Fried, Gerd A. Kullak-Ublick, Stephan R. Vavricka, Benjamin Misselwitz, Klaus Seuwen, Carsten A. Wagner, Jyrki J.
    Cellular and Molecular Gastroenterology and Hepatology.2016; 2(6): 796.     CrossRef
  • Bile acids destabilise HIF-1α and promote anti-tumour phenotypes in cancer cell models
    J. P. Phelan, F. J. Reen, N. Dunphy, R. O’Connor, F. O’Gara
    BMC Cancer.2016;[Epub]     CrossRef
  • Better Living through Chemistry: Caloric Restriction (CR) and CR Mimetics Alter Genome Function to Promote Increased Health and Lifespan
    Zoe E. Gillespie, Joshua Pickering, Christopher H. Eskiw
    Frontiers in Genetics.2016;[Epub]     CrossRef
  • Folic Acid Represses Hypoxia-Induced Inflammation in THP-1 Cells through Inhibition of the PI3K/Akt/HIF-1α Pathway
    Xiaoyan Huang, Zhiying He, Xinwei Jiang, Mengjun Hou, Zhihong Tang, Xiaozhou Zhen, Yuming Liang, Jing Ma, George Simos
    PLOS ONE.2016; 11(3): e0151553.     CrossRef
  • Cinnamic aldehyde suppresses hypoxia-induced angiogenesis via inhibition of hypoxia-inducible factor-1α expression during tumor progression
    Woom-Yee Bae, Jae-Sun Choi, Ja-Eun Kim, Joo-Won Jeong
    Biochemical Pharmacology.2015; 98(1): 41.     CrossRef
  • Clinicopathological Significance of MicroRNA-20b Expression in Hepatocellular Carcinoma and Regulation of HIF-1αand VEGF Effect on Cell Biological Behaviour
    Tong-min Xue, Li-de Tao, Miao Zhang, Jie Zhang, Xia Liu, Guo-feng Chen, Yi-jia Zhu, Pei-Jian Zhang
    Disease Markers.2015; 2015: 1.     CrossRef
  • HIF-1α Contributes to Proliferation and Invasiveness of Neuroblastoma Cells via SHH Signaling
    Sheng Chen, Min Zhang, Lili Xing, Yue Wang, Yongtao Xiao, Yeming Wu, Sharmila Shankar
    PLOS ONE.2015; 10(3): e0121115.     CrossRef
  • Autoacetylation regulates differentially the roles of ARD1 variants in tumorigenesis
    JI HAE SEO, JI-HYEON PARK, EUN JI LEE, TAM THUY LU VO, HOON CHOI, JAE KYUNG JANG, HEE-JUN WEE, BUM JU AHN, JONG-HO CHA, MIN WOOK SHIN, KYU-WON KIM
    International Journal of Oncology.2015; 46(1): 99.     CrossRef
  • Transcriptome sequencing revealed differences in the response of renal cancer cells to hypoxia and CoCl2 treatment
    Nadezhda Zhigalova, Artem Artemov, Alexander M. Mazur, Egor B. Prokhortchouk
    F1000Research.2015; 4: 1518.     CrossRef
  • The Role of Hypoxia Inducible Factor-1 in Hepatocellular Carcinoma
    Dongjun Luo, Zhongxia Wang, Junyi Wu, Chunping Jiang, Junhua Wu
    BioMed Research International.2014; 2014: 1.     CrossRef
  • Epidithiodiketopiperazines (ETPs) exhibit in vitro antiangiogenic and in vivo antitumor activity by disrupting the HIF-1α/p300 complex in a preclinical model of prostate cancer
    Kelie M Reece, Emily D Richardson, Kristina M Cook, Tessa J Campbell, Stephen T Pisle, Alesia J Holly, David J Venzon, David J Liewehr, Cindy H Chau, Douglas K Price, William D Figg
    Molecular Cancer.2014;[Epub]     CrossRef
  • Exploring the Modulation of Hypoxia-Inducible Factor (HIF)-1α by Volatile Anesthetics as a Possible Mechanism Underlying Volatile Anesthetic-Induced CNS Injury
    Emma K. Giles, Andrew J. Lawrence, Jhodie R. Duncan
    Neurochemical Research.2014; 39(9): 1640.     CrossRef
  • Activation-induced necroptosis contributes to B-cell lymphopenia in active systemic lupus erythematosus
    H Fan, F Liu, G Dong, D Ren, Y Xu, J Dou, T Wang, L Sun, Y Hou
    Cell Death & Disease.2014; 5(9): e1416.     CrossRef
  • Sleep apnoea and cancer: the new challenge
    Patrick Lévy, Diane Godin-Ribuot, Jean-Louis Pepin
    European Respiratory Journal.2014; 43(6): 1567.     CrossRef
  • [18F]Fluorodeoxyglucose accumulation as a biological marker of hypoxic status but not glucose transport ability in gastric cancer
    Ryusuke Takebayashi, Kunihiko Izuishi, Yuka Yamamoto, Reiko Kameyama, Hirohito Mori, Tsutomu Masaki, Yasuyuki Suzuki
    Journal of Experimental & Clinical Cancer Research.2013;[Epub]     CrossRef
  • The influence of pH and hypoxia on tumor metastasis
    Mariam Abaza, Yunus A Luqmani
    Expert Review of Anticancer Therapy.2013; 13(10): 1229.     CrossRef
  • Recent advances in hypoxia-inducible factor (HIF)-1 inhibitors
    Yan Xia, Hyun-Kyung Choi, Kyeong Lee
    European Journal of Medicinal Chemistry.2012; 49: 24.     CrossRef
  • Molecular Mechanism Underlying the Detection of Colorectal Cancer by 18F-2-Fluoro-2-Deoxy-d-Glucose Positron Emission Tomography
    Kunihiko Izuishi, Yuka Yamamoto, Takanori Sano, Ryusuke Takebayashi, Yoshihiro Nishiyama, Hirohito Mori, Tsutomu Masaki, Asahiro Morishita, Yasuyuki Suzuki
    Journal of Gastrointestinal Surgery.2012; 16(2): 394.     CrossRef
  • A hypoxia-dependent upregulation of hypoxia-inducible factor-1 by nuclear factor-κB promotes gastric tumour growth and angiogenesis
    S Y Nam, Y S Ko, J Jung, J Yoon, Y H Kim, Y J Choi, J W Park, M S Chang, W H Kim, B L Lee
    British Journal of Cancer.2011; 104(1): 166.     CrossRef
  • Constitutive phosphorylation of the FOXO1 transcription factor in gastric cancer cells correlates with microvessel area and the expressions of angiogenesis-related molecules
    Sue Youn Kim, Jiyeon Yoon, Young San Ko, Mee Soo Chang, Jong-Wan Park, Hee Eun Lee, Min A Kim, Ji Hun Kim, Woo Ho Kim, Byung Lan Lee
    BMC Cancer.2011;[Epub]     CrossRef
  • Terpestacin Inhibits Tumor Angiogenesis by Targeting UQCRB of Mitochondrial Complex III and Suppressing Hypoxia-induced Reactive Oxygen Species Production and Cellular Oxygen Sensing
    Hye Jin Jung, Joong Sup Shim, Jiyong Lee, Young Mi Song, Ki Chung Park, Seung Hoon Choi, Nam Doo Kim, Jeong Hyeok Yoon, Paul T. Mungai, Paul T. Schumacker, Ho Jeong Kwon
    Journal of Biological Chemistry.2010; 285(15): 11584.     CrossRef
  • The Clinicopathological Significance of Tissue Levels of Hypoxia-Inducible Factor-1α and Vascular Endothelial Growth Factor in Gastric Cancer
    Seong-Eun Kim, Ki-Nam Shim, Sung-Ae Jung, Kwon Yoo, Joo Ho Lee
    Gut and Liver.2009; 3(2): 88.     CrossRef
  • 29,963 View
  • 622 Download
  • 77 Crossref
Close layer
Original Articles
The Effects of Insulin-like Growth Factor-I (IGF-I) in Mouse Lung Cancer Cells
Seung Min Kwak, Se Kyu Kim, Sung Kyu Kim, Chul Ho Cho
Cancer Res Treat. 2002;34(2):122-127.   Published online April 30, 2002
DOI: https://doi.org/10.4143/crt.2002.34.2.122
AbstractAbstract PDF
PURPOSE
Insulin-like growth factor-I (IGF-I) is an important mitogen in many types of malignancies. The purpose of this study was to evaluate the role of the IGF system on cell proliferation and cell death in mouse lung cancer cell lines (3LL).
MATERIALS AND METHODS
Northern analysis was performed in 3LL cells. We evaluated the phosphorylation of IGF-I receptor (IGF-IR) with IGF-I stimulation. MTT assay was performed after treating 3LL cells with IGF-I and the treatment effect on cell death in the presence of anticancer drug was investigated.
RESULTS
Northern analysis revealed the presence of IGF-I and IGF-IR mRNA expression in 3LL cells. IGF-I increased cellular proliferation in serum free media. IGF-I also stimulated the tyrosine phosphorylation of two proteins: one, with a molecular mass of 95 kDa, was the beta-subunit of IGF-IR; the other, with an approximate molecular mass of 185 kDa, was originally identified as the insulin receptor substrate-I (IRS-I). IGF-I at a low concentration inhibited the cell death induced by adriamycin.
CONCLUSION
IGF-I, a mitogen through the phosphorylation of the IGF-IR beta-subunit, acts as a survival factor to inhibit cell death. Therefore, these findings suggest that IGF-I and IGF-IR are involved in both the cell proliferation and cell death associated with cancer cell growth.
  • 3,834 View
  • 23 Download
Close layer
Apoptosis in Renal Cell Carcinoma: Correlation to Apoptosis Related Genes and Cell Proliferation, and Its Prognostic Significance
Ji Shin Lee, Jong Jae Jung, Sung Taek Lee, Chang Soo Park
J Korean Cancer Assoc. 2001;33(1):9-15.
AbstractAbstract PDF
PURPOSE
To investigate the prognostic role of apoptosis and to evaluate the relationship between apoptosis and apoptosis-related genes, as well as cell proliferation in renal cell carcinoma (RCC).
MATERIALS AND METHODS
Apoptosis was detected by using the terminal deoxynucleotidyl transferase (TdT) mediated dUTP nick-end labeling (TUNEL) technique in 67 formalin-fixed and paraffin-embedded RCC specimens. Immunohistochemical stainings for p53 and retinoblastoma (Rb) proteins and proliferating cell nuclear antigen (PCNA) were also conducted simultaneously.
RESULTS
The apoptotic index (AI) varied from 0.2% to 25.5%. The PCNA index (PI) ranged from 2.1% to 70.3%. The expression of p53 protein was found in 31 of 67 (46.3%) cases. Abnormal expression of Rb was seen in 23 of 67 (34.3%) cases. There was a statistically significant positive correlation between AI and increasingnuclear grade (p<0.001). A significant correlation was found between AI and PI (r=0.329, p<0.01). When comparing the AI with the expression of p53 and Rb proteins, there was no significant difference. In univariate survival analysis, nuclear grade, TNM stage, PI, expression of Rb and AI were significantly associated with shortened survival. However, TNM stage was the only independent prognostic factors by multivariate analysis.
CONCLUSION
The present findings indicate that apoptosis in RCC is closely associated with cell proliferation, but not with the expression of p53 and Rb proteins. In multivariate analysis, the AI does not carry an independent prognostic significance.
  • 2,747 View
  • 27 Download
Close layer
Reduced Expression of p27 Correlates with High Grade Malignancy and is Associated with Poor Clinical Outcome in Human Greast Cancer
Se Hwan Han, Hong Yong Kim, Kyeong Mee Park, Myeong Soo Lee, Hong Joo Kim, Young Doug Kim, Hyun Suk Suh
J Korean Cancer Assoc. 1999;31(3):492-498.
AbstractAbstract PDF
PURPOSE
The cyclin-dependent kinase inhibitor p27Kipl is a negative regulator of cell proliferation. Its expression is known to be altered in proteasome-dependent manner without changes in DNA level. Reduced expression of p27Kipl is associated with aggressive behavior in a variety of human cancers. We investigated expression of p27Kipl protein in human breast cancer using immunohistochemistry to assess its biologic implication along with cell cycle analysis by flow cytometry.
MATERIALS AND METHODS
We peformed immunohistochemical assay for expression of p27 in total 68 patients with invasive ductal cancer along with 27 benign ductal hyperplasia and 10 ductal carcinoma in situ. The data were analyzed in association with proliferative activity of the same tissues and biologic parameters.
RESULTS
In epithelial cells of normal and benign breast disease, expression of p27Kipl was well preserved while its expression markedly decreased in breast cancer (45 of 68). Expression of p27Kipl was significantly reduced in poorly differentiated cancers and in advanced stage of the disease. Levels of p27Kipl expression correlated with cell populations in GO/Gl phase of the cell cycle. In survival analysis, p27Kipl was useful to predict disease-free survival but not overall survival of the patients after adjuvant chemotherapy.
CONCLUSION
p27Kipl seems to have a role in cell proliferation and differentiation process during carcinogenesis of breast cancer. The results of present study suggest that p27Kipl can be used in predicting response to systemic chemotherapy in a subset of patients with breast cancer.
  • 2,756 View
  • 14 Download
Close layer
Prognostic Significance of p53 Overexpression in Gastric Carcinoma
Yong Il Kim, Jin Pok Kim, Woo Ho Kim, Sang Yong Song, Chong Jai Kim
J Korean Cancer Assoc. 1994;26(5):702-711.
AbstractAbstract PDF
The overexpession of p53 protein was immunohistochemically studied in a total of 92 gastric carcinomas, of which 40 cases(43.5%) showed nuclear immunoreactivity. Older age(p<0.001), lymph node metastasis(p<0.05), stage(p<0.05), and higher expression of proliferating cell nuclear antigen(p<0.05) were correlated with p53 overexpression. Other clinicopathologic parameters including sex, tumor site, WHO classification, Ming classification, inflammation degree, desmoplasia degree, tumor invasion depth, gross type, size, and recurrence were not related with p53 overexpression. However, intestinal type(56.3%) by Lauren classification and advanced gastric carcinoma(48.0%) showed higher incidence of p53 overexpression than diffuse type(36.7%) and early gastric carcinoma(23.5%), respectively(0.05< p<0.1). Lifetest showed that p53 overexpression was not related with patients survival, although tumor site, size, depth of invasion, stage, lymph node metastasis, and recurrence were closely correlated with patients survivaL We concluded that p53 overexpression was a common event in gastric carcinogenesis and may play a role not in late, invasive stage of tumor progression related with patient survival, but in early, proliferative stage of gastric carcioma progression.
  • 2,636 View
  • 14 Download
Close layer

Cancer Res Treat : Cancer Research and Treatment
Close layer
TOP