1
|
Chen M, Zhu H, Li J, Luo D, Zhang J, Liu W, Wang J. Research progress on the relationship between AURKA and tumorigenesis: the neglected nuclear function of AURKA. Ann Med 2024; 56:2282184. [PMID: 38738386 PMCID: PMC11095293 DOI: 10.1080/07853890.2023.2282184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/31/2023] [Indexed: 05/14/2024] Open
Abstract
AURKA is a threonine or serine kinase that needs to be activated by TPX2, Bora and other factors. AURKA is located on chromosome 20 and is amplified or overexpressed in many human cancers, such as breast cancer. AURKA regulates some basic cellular processes, and this regulation is realized via the phosphorylation of downstream substrates. AURKA can function in either the cytoplasm or the nucleus. It can promote the transcription and expression of oncogenes together with other transcription factors in the nucleus, including FoxM1, C-Myc, and NF-κB. In addition, it also sustains carcinogenic signaling, such as N-Myc and Wnt signaling. This article will focus on the role of AURKA in the nucleus and its carcinogenic characteristics that are independent of its kinase activity to provide a theoretical explanation for mechanisms of resistance to kinase inhibitors and a reference for future research on targeted inhibitors.
Collapse
Affiliation(s)
- Menghua Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huijun Zhu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jian Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Danjing Luo
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jiaming Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenqi Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jue Wang
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Gao W, Shen R. Nanogel enhances the efficacy of MLN8237 in treating hepatocellular carcinoma. J Biomater Appl 2023; 38:527-537. [PMID: 37695622 DOI: 10.1177/08853282231202326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
MLN8237, a specific inhibitor of Aurora-A kinase, is proved to be a potential treatment strategy for hepatocellular carcinoma (HCC). Nanogels improve the efficacy of doxorubicin. Therefore, this study aims to investigate the increase in the effect of nanogels on MLN8237 in inhibiting HCC. Doxorubicin or MLN8237 was used as an anti-tumor drug models which were packaged by organic solvent volatilization method to obtain the doxorubicin-loaded nanogel and the MLN8237-loaded nanogel. Subsequently, CCK8 assay, cell cycle assay, apoptosis assay, real-time PCR, western blotting assay and animal experiments were used to detect the effects of MLN8237 nanogel on the proliferation, cell cycle, apoptosis, tumor growth, mRNA and protein levels of aurora-A and PUMA, and AKT phosphorylation levels in HCC cell lines. The results show that the nanogels can realize pH-regulated hydrophobicity reversal, have certain stability, and can realize lysosomal escape. Moreover, the MLN8237-loaded nanogel has a stronger ability to inhibit HCC cell proliferation, block cell cycle, promote apoptosis and inhibit tumor growth than free MLN8237 by suppressing aurora-A and AKT phosphorylation. In short, nanogel can enhance the efficacy of MLN8237.
Collapse
Affiliation(s)
- Wei Gao
- Department of General Surgery, The Second People's Hospital of Tongxiang, Zhejiang, China
| | - Rongxing Shen
- Department of General Surgery, The Second People's Hospital of Tongxiang, Zhejiang, China
| |
Collapse
|
3
|
Fan L, Huang X, Chen J, Zhang K, Gu YH, Sun J, Cui SY. Long Noncoding RNA MALAT1 Contributes to Sorafenib Resistance by Targeting miR-140-5p/Aurora-A Signaling in Hepatocellular Carcinoma. Mol Cancer Ther 2020; 19:1197-1209. [PMID: 32220970 DOI: 10.1158/1535-7163.mct-19-0203] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 09/09/2019] [Accepted: 03/11/2020] [Indexed: 11/16/2022]
Abstract
Long noncoding RNAs (lncRNA) have been found to play critical roles in tumorigenesis and the development of various cancers, including hepatocellular carcinoma (HCC). Metastasis associated with lung adenocarcinoma transcript-1 (MALAT1) has been identified as an oncogene and prognostic biomarker in HCC. Here, we demonstrated that MALAT1 expression was obviously high in sorafenib-resistant HCC cells. Furthermore, knockdown of MALAT1 increased sorafenib sensitivity in nonresponsive HCC cells, whereas forced expression of MALAT1 conferred sorafenib resistance to responsive HCC cells in vitro In addition, loss/gain-of-function assays revealed that MALAT1 promoted cell proliferation, migration, and epithelial-mesenchymal transition in HCC cells. Mechanistically, MALAT1 regulated Aurora-A expression by sponging miR-140-5p, thus promoting sorafenib resistance in HCC cells. Moreover, MALAT1 inhibition enhanced the antitumor efficacy of sorafenib in vivo Clinically, we found that MALAT1 expression was negatively correlated with miR-140-5p expression but positively correlated with Aurora-A expression in patients with HCC and that upregulated MALAT1 was closely correlated with poor survival outcomes in patients with HCC. These findings indicated that MALAT1 may be a novel target for prognosis prediction and therapeutic strategies in patients with HCC treated with sorafenib.
Collapse
Affiliation(s)
- Lei Fan
- Department of General Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nangjing, P.R. China
| | - Xiang Huang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jing Chen
- Department of Respiratory, Zhongda Hospital, Southeast University, Nanjing, P.R. China
| | - Kai Zhang
- Department of Respiratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yan-Hong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jing Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China.
| | - Shi-Yun Cui
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China.
| |
Collapse
|
4
|
Weidle UH, Schmid D, Birzele F, Brinkmann U. MicroRNAs Involved in Metastasis of Hepatocellular Carcinoma: Target Candidates, Functionality and Efficacy in Animal Models and Prognostic Relevance. Cancer Genomics Proteomics 2020; 17:1-21. [PMID: 31882547 PMCID: PMC6937123 DOI: 10.21873/cgp.20163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is responsible for the second-leading cancer-related death toll worldwide. Although sorafenib and levantinib as frontline therapy and regorafenib, cabazantinib and ramicurimab have now been approved for second-line therapy, the therapeutic benefit is in the range of only a few months with respect to prolongation of survival. Aggressiveness of HCC is mediated by metastasis. Intrahepatic metastases and distant metastasis to the lungs, lymph nodes, bones, omentum, adrenal gland and brain have been observed. Therefore, the identification of metastasis-related new targets and treatment modalities is of paramount importance. In this review, we focus on metastasis-related microRNAs (miRs) as therapeutic targets for HCC. We describe miRs which mediate or repress HCC metastasis in mouse xenograft models. We discuss 18 metastasis-promoting miRs and 35 metastasis-inhibiting miRs according to the criteria as outlined. Six of the metastasis-promoting miRs (miR-29a, -219-5p, -331-3p, 425-5p, -487a and -1247-3p) are associated with unfavourable clinical prognosis. Another set of six down-regulated miRs (miR-101, -129-3p, -137, -149, -503, and -630) correlate with a worse clinical prognosis. We discuss the corresponding metastasis-related targets as well as their potential as therapeutic modalities for treatment of HCC-related metastasis. A subset of up-regulated miRs -29a, -219-5p and -425-5p and down-regulated miRs -129-3p and -630 were evaluated in orthotopic metastasis-related models which are suitable to mimic HCC-related metastasis. Those miRNAs may represent prioritized targets emerging from our survey.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Daniela Schmid
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| | - Fabian Birzele
- Pharmaceutical Sciences, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| | - Ulrich Brinkmann
- Large Molecule Research, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
5
|
Shen ZT, Chen Y, Huang GC, Zhu XX, Wang R, Chen LB. Aurora-a confers radioresistance in human hepatocellular carcinoma by activating NF-κB signaling pathway. BMC Cancer 2019; 19:1075. [PMID: 31703572 PMCID: PMC6842208 DOI: 10.1186/s12885-019-6312-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 10/30/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Radiotherapy failure is a significant clinical challenge due to the development of resistance in the course of treatment. Therefore, it is necessary to further study the radiation resistance mechanism of HCC. In our early study, we have showed that the expression of Aurora-A mRNA was upregulated in HCC tissue samples or cells, and Aurora-A promoted the malignant phenotype of HCC cells. However, the effect of Aurora-A on the development of HCC radioresistance is not well known. METHODS In this study, colony formation assay, MTT assays, flow cytometry assays, RT-PCR assays, Western blot, and tumor xenografts experiments were used to identify Aurora-A promotes the radioresistance of HCC cells by decreasing IR-induced apoptosis in vitro and in vivo. Dual-luciferase reporter assay, MTT assays, flow cytometry assays, and Western blot assay were performed to show the interactions of Aurora-A and NF-κB. RESULTS We established radioresistance HCC cell lines (HepG2-R) and found that Aurora-A was significantly upregulated in those radioresistant HCC cells in comparison with their parental HCC cells. Knockdown of Aurora-A increased radiosensitivity of radioresistant HCC cells both in vivo and in vitro by enhancing irradiation-induced apoptosis, while upregulation of Aurora-A decreased radiosensitivity by reducing irradiation-induced apoptosis of parental cells. In addition, we have showed that Aurora-A could promote the expression of nuclear IkappaB-alpha (IκBα) protein while enhancing the activity of NF-kappaB (κB), thereby promoted expression of NF-κB pathway downstream effectors, including proteins (Mcl-1, Bcl-2, PARP, and caspase-3), all of which are associated with apoptosis. CONCLUSIONS Aurora-A reduces radiotherapy-induced apoptosis by activating NF-κB signaling, thereby contributing to HCC radioresistance. Our results provided the first evidence that Aurora-A was essential for radioresistance in HCC and targeting this molecular would be a potential strategy for radiosensitization in HCC.
Collapse
Affiliation(s)
- Ze-Tian Shen
- Department of Radiation Oncology, Jinling Hospital, Nanjing Medical School University, Nanjing, Jiangsu, China
| | - Ying Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Gui-Chun Huang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China
| | - Xi-Xu Zhu
- Department of Radiation Oncology, Jinling Hospital, Nanjing Medical School University, Nanjing, Jiangsu, China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu, China.
| | - Long-Bang Chen
- Department of Medical Oncology, Jinling Hospital, Nanjing Medical School University, Nanjing, Jiangsu, China.
| |
Collapse
|
6
|
A Novel Aurora-A Inhibitor (MLN8237) Synergistically Enhances the Antitumor Activity of Sorafenib in Hepatocellular Carcinoma. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 13:176-188. [PMID: 30292139 PMCID: PMC6172479 DOI: 10.1016/j.omtn.2018.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/07/2018] [Accepted: 08/19/2018] [Indexed: 02/08/2023]
Abstract
Currently, sorafenib-based therapy is the standard treatment for advanced hepatocellular carcinoma (HCC), and there is a strong rationale for investigating its use in combination with other agents to achieve better therapeutic effects. Aurora-A, a member of a family of mitotic serine/threonine kinases, is frequently overexpressed in human cancers and therefore represents a target for therapy. Here, we investigated a novel Aurora-A inhibitor, MLN8237, together with sorafenib in HCC cells in vitro and in vivo, and elucidated the possible molecular mechanism. Here, it was found that MLN8237 was strongly synergistic with sorafenib in inhibition of HCC progression by altering cell growth, cell-cycle regulation, apoptosis, migration, invasion, and angiogenesis. Mechanism dissection suggests that the combination of MLN8237 and sorafenib led to significant inhibition of the activation of phospho-Akt (p-Akt) and phospho-p38 mitogen-activated protein kinase (p-p38 MAPK) and their downstream genes including CDK4, cyclinD1, and VEGFA. The activators of p-Akt and p-p38 MAPK signaling partially reversed the synergistic inhibitory effects of sorafenib and MLN8237 on HCC progression. Subsequent in vivo studies further confirmed the synergistic effects of sorafenib and MLN8237. Collectively, the newly developed sorafenib-MLN8237 combination may be a novel therapy to better inhibit HCC progression.
Collapse
|
7
|
Cui S, Zhang K, Li C, Chen J, Pan Y, Feng B, Lu L, Zhu Z, Wang R, Chen L. Methylation-associated silencing of microRNA-129-3p promotes epithelial-mesenchymal transition, invasion and metastasis of hepatocelluar cancer by targeting Aurora-A. Oncotarget 2018; 7:78009-78028. [PMID: 27793005 PMCID: PMC5363640 DOI: 10.18632/oncotarget.12870] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/14/2016] [Indexed: 12/12/2022] Open
Abstract
Metastasis and recurrence has become one major obstacle for further improving the survival of hepatocelluar cancer (HCC) patients. Therefore, it is critical to elucidate the mechanisms involved in HCC metastasis. This study aimed to investigate the roles of microRNA (miR)-129-3p in HCC metastasis and its possible molecular mechanisms. By using microarray analysis to compare levels of different miRNAs in HCC tissues with or without lymph node metastasis (LNM), we showed that HCC tissues with LNM had reduced levels of miR-129-3p, which was related to its promoter hypermethylation and correlated with tumor metastasis, recurrence and poor prognosis. Gain - and loss - of - function assays indicated that re-expression of miR-129-3p could reverse epithelial-mesenchymal transition (EMT), and reduce in vitro invasion and in vivo metastasis of HCC cells. Aurora-A, a serine/threonine protein kinase, was identified as a direct target of miR-129-3p. Knockdown of Aurora-A phenocopied the effect of miR-129-3p overexpression on HCC metastasis. In addition, Aurora-A upregulation could partially rescue the effect of miR-129-3p. We further demonstrated that activation of PI3K/Akt and p38-MAPK signalings were involved in miR-129-3p-mediated HCC metastasis. These findings suggest that methylation-mediated miR-129-3p downregulation promotes EMT, in vitro invasion and in vivo metastasis of HCC cells via activation of PI3K/Akt and p38-MAPK signalings partially by targeting Aurora-A. Therefore, miR-129-3p may be a novel prognostic biomarker and potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Shiyun Cui
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Kai Zhang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Chen Li
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Jing Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Yan Pan
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Bing Feng
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Lei Lu
- Liver Disease Center of PLA, The 81th Hospital of PLA, Nanjing 210002, Jiangsu, PR China
| | - Ziman Zhu
- Department of Hepatobiliary Surgery, First Hospital Affiliated to the Chinese PLA General Hospital, Haidian District, Beijing 100048, PR China
| | - Rui Wang
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| | - Longbang Chen
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210002, Jiangsu, PR China
| |
Collapse
|
8
|
Bao Z, Lu L, Liu X, Guo B, Zhai Y, Li Y, Wang Y, Xie B, Ren Q, Cao P, Han Y, Jia W, Chen M, Liang X, Wang X, Zeng YX, He F, Zhang H, Cui Y, Zhou G. Association between the functional polymorphism Ile31Phe in the AURKA gene and susceptibility of hepatocellular carcinoma in chronic hepatitis B virus carriers. Oncotarget 2017; 8:54904-54912. [PMID: 28903390 PMCID: PMC5589629 DOI: 10.18632/oncotarget.18613] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/22/2017] [Indexed: 11/25/2022] Open
Abstract
Aurora kinase A (AURKA) is a serine threonine kinase which affects chromosomal separation and mitotic spindle stability through interaction with the centrosome during mitosis. Two functional nonsynonymous polymorphisms of the AURKA gene (Ile31Phe and Val57Ile) have been reported recently. We analyzed the association between the two polymorphisms and risk of the occurrence of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) in the Guangxi population consisting of 348 patients with HCC and 359 control subjects, and then validated the significant association in the Guangdong population consisting of 440 cases and 456 controls. All of the participants were of Chinese origin and HBV carriers. The two polymorphisms were genotyped by polymerase chain reaction-restriction fragment length polymorphism assay or Sequenom MassARRAY iPLEX platform. In the Guangxi population, carriers of the AURKA 31Phe allele (Ile/Phe + Phe/Phe) were significantly associated with decreased susceptibility to HBV-related HCC when compared with noncarriers (Ile/Ile) (odds ratio [OR] = 0.63, 95% confidence interval [CI] = 0.46-0.86, P = 3.4 × 10-3). On the contrary, no significant association was found between Val57Ile and HBV-related HCC occurrence. The association of Ile31Phe with HBV-related HCC occurrence was confirmed in the Guangdong population (OR = 0.64, 95% CI = 0.49-0.83, P = 8.0 × 10-4). The pooled analysis gave a joint P value of 5.5 × 10-6 (joint OR = 0.63, 95% CI = 0.52-0.77). Our findings suggest that AURKA Ile31Phe may play a role in mediating the susceptibility to HBV-related HCC among Chinese.
Collapse
Affiliation(s)
- Zhiyu Bao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,Guangxi Medical University, Nanning, China.,Affiliated Hospital of Jining Medical University, Jining, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Lei Lu
- Department of Surgical Oncology, Bayi Hospital Affiliated Nanjing University of Chinese Medicine, Jindu Hospital, Nanjing, China
| | - Xinyi Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Bingqian Guo
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Yun Zhai
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Yuanfeng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Yahui Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Bobo Xie
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Qian Ren
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Pengbo Cao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Yuqing Han
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Weihua Jia
- State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Minshan Chen
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Xuan Wang
- Department of Surgical Oncology, Bayi Hospital Affiliated Nanjing University of Chinese Medicine, Jindu Hospital, Nanjing, China
| | - Yi-Xin Zeng
- State Key Laboratory of Oncology in Southern China, Guangzhou, China.,Department of Experimental Research, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Hongxing Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China
| | - Ying Cui
- Guangxi Medical University, Nanning, China
| | - Gangqiao Zhou
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China.,National Engineering Research Center for Protein Drugs, Beijing, China.,National Center for Protein Sciences Beijing, Beijing, China.,Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
9
|
Wu J, Yang L, Shan Y, Cai C, Wang S, Zhang H. AURKA promotes cell migration and invasion of head and neck squamous cell carcinoma through regulation of the AURKA/Akt/FAK signaling pathway. Oncol Lett 2016; 11:1889-1894. [PMID: 26998095 DOI: 10.3892/ol.2016.4110] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 12/10/2015] [Indexed: 12/15/2022] Open
Abstract
The present study aimed to investigate the mechanism by which Aurora kinase A (AURKA) promotes cell migration and invasion in head and neck squamous cell carcinoma (HNSCC). Transwell assays were performed to investigate the cell migration and invasion abilities of AURKA, whilst western blotting was used to analyze the protein expression in FaDu and Hep2 cells, each treated with pharmacological inhibitors. Following the inhibition of AURKA, Akt and focal adhesion kinase (FAK), the migration and invasion of the FaDu and Hep2 cells decreased. The expression of phosphorylated (p)-AURKA and p-FAK (Y397) was observed to decrease following FaDu and Hep2 cell treatment with VX-680, a small molecular inhibitor of AURKA. The expression of p-Akt and p-FAK (Y397) ceased following treatment with the Akt inhibitor triciribine. The expression of p-FAK (Y397) decreased, however, p-Akt expression did not change following treatment with the FAK inhibitor TAE226. In conclusion, AURKA activates FAK through the AURKA/Akt/FAK signaling pathway, promoting the migration and invasion of HNSCC cells, which may subsequently provide a novel approach for the treatment of HNSCC.
Collapse
Affiliation(s)
- Jichang Wu
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Liyun Yang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Yamin Shan
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Changping Cai
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Shili Wang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Hao Zhang
- Department of Otolaryngology, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| |
Collapse
|
10
|
Aurora-A promotes chemoresistance in hepatocelluar carcinoma by targeting NF-kappaB/microRNA-21/PTEN signaling pathway. Oncotarget 2015; 5:12916-35. [PMID: 25428915 PMCID: PMC4350360 DOI: 10.18632/oncotarget.2682] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/04/2014] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is highly resistant to chemotherapy. Previously, we have shown that Aurora-A mRNA is upregulated in HCC cells or tissues and silencing of Aurora-A using small interfering RNA (siRNA) decreases growth and enhances apoptosis in HCC cells. However, the clinical significance of Aurora-A protein expression in HCC and association between Aurora-A expression and HCC chemoresistance is unclear. Here, we showed that Aurora-A protein is upregulated in HCC tissues and significantly correlated with recurrence-free and overall survival of patients and multivariate analysis indicated that immunostaining of Aurora-A will be an independent prognostic factor for patients. Silencing of Aurora-A significantly increased the chemosensitivity of HCC cells both in vitro and in vivo, while overexpression of Aurora-A induced the opposite effects. Furthermore, overexpression of Aurora-A reduces chemotherapy-induced apoptosis by promoting microRNA-21 expression, which negatively regulates PTEN and then inhibits caspase-3-mediated apoptosis induction. Mechanically, we demonstrated that Aurora-A promotes expression of nuclear Ikappaβ-alpha (Iκβα) protein and enhances NF-kappa B (NF-κB) activity, thus promotes the transcription of miR-21. This study first reported the involvement of Aurora-A/NF-κB/miR-21/PTEN/Akt signaling axis in chemoresistance of HCC cells, suggesting that targeting this signaling pathway would be helpful as a therapeutic strategy for the reversal of chemoresistance in HCC.
Collapse
|
11
|
He Y, Jiang W, Qian X, Liu F, Zhang Q, You C. Role of Aurora-A in Ovarian Cancer: A Meta-Analysis. Oncol Res Treat 2015; 38:442-7. [PMID: 26406304 DOI: 10.1159/000439194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/29/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Recently, several studies have examined associations between Aurora-A expression and clinical outcome in patients with ovarian cancer, but yielded conflicting results with respect to survival. Therefore, the aim of this study was to evaluate the prognostic significance of Aurora-A in ovarian cancer by performing a meta-analysis. METHODS PubMed, Cochrane library, Web of Science, Embase, Medline and Chinese BioMed Database (CBM) databases were searched systematically and only articles in which Aurora-A expression was detected by immunohistochemical staining were included. Hazard ratios (HRs) with 95% confidence intervals (CIs) were extracted and pooled for overall survival (OS) and disease-free survival (DFS). RESULTS Our results show that the pooled HR for OS was 1.40 (95% CI 0.82-1.98, p < 0.01) by univariate analysis in 7 articles (1,028) and 0.32 (95% CI 0.04-0.615, p = 0.23) by multivariate analysis in 3 studies (155). The association between Aurora-A expression and DFS was also statistically significant in 5 studies (HR = 1.14, 95% CI 0.50-1.78, p < 0.01). CONCLUSION This present meta-analysis suggests that the Aurora-A expression may be associated with poor prognosis in patients with ovarian cancer. Furthermore, studies of larger scale and well-matched regimes are warranted to confirm the findings in the future.
Collapse
Affiliation(s)
- Yuan He
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
12
|
Zhang J, Li B, Yang Q, Zhang P, Wang H. Prognostic value of Aurora kinase A (AURKA) expression among solid tumor patients: a systematic review and meta-analysis. Jpn J Clin Oncol 2015; 45:629-36. [DOI: 10.1093/jjco/hyv058] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/29/2015] [Indexed: 12/16/2022] Open
|
13
|
AURKA is a predictor of chemotherapy response and prognosis for patients with advanced oral squamous cell carcinoma. Tumour Biol 2014; 36:3557-64. [PMID: 25547434 DOI: 10.1007/s13277-014-2992-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/17/2014] [Indexed: 10/24/2022] Open
Abstract
Increasing evidence proposes the benefits of cisplatin-based chemotherapy in a subpopulation of patients with oral squamous cell carcinoma (OSCC), yet reliable indicators for this subpopulation are poorly explored. AURKA, also known as aurora kinase A, playing important functions in cell mitosis and making cells resistant to cisplatin through dysregulation of DNA damage repair networks, has been reported to be upregulated in OSCC, making AURKA a promising indicator. In this study, we recruited 78 patients with advanced OSCC to examine the expression of AURKA and the correlation with chemotherapy response and clinical outcomes. We found that AURKA was strongly expressed in 31 (39.74 %) of the 78 advanced OSCC samples and its expression was significantly associated with cisplatin resistance (P = 0.023), clinical recurrence (P = 0.021), and 5-year survival (P = 0.019). Chemotherapy increased AURKA expression in post-chemotherapy samples, yet with no significance (P = 0.101). Multivariate Cox proportional hazards regression model analysis demonstrated that lymph node samples with positive, strong AURKA staining, and poor chemotherapy response were independently associated with the 5-year survival and disease-free survival. Inhibiting AURKA expression in OSCC cell lines remarkably increased their sensitivity to cisplatin treatment by 2.5-fold difference. Our results imply that the overexpression of AURKA in advanced OSCC not only plays a role in the disease course but also shows an involvement in cisplatin treatment response. AURKA level may be a valuable predictor for patients with advanced OSCC, with downregulation of AURKA being a promising adjuvant therapy in this patient population.
Collapse
|
14
|
Cui SY, Huang JY, Chen YT, Song HZ, Huang GC, De W, Wang R, Chen LB. The role of Aurora A in hypoxia-inducible factor 1α-promoting malignant phenotypes of hepatocelluar carcinoma. Cell Cycle 2013; 12:2849-66. [PMID: 23966163 DOI: 10.4161/cc.25916] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Overexpression of both hypoxia-inducible factor 1α (HIF-1α) and Aurora A has been found in hepatocellular carcinoma (HCC). However, whether HIF-1α and Aurora A synergistically promote malignant phenotypes of HCC cells is unknown. The purpose of this study was to investigate the roles and functional correlation of HIF-1α and Aurora A in HCC progression. Immunohistochemistry was performed to detect HIF-1α and Aurora A protein expression in 55 primary HCC and corresponding non-tumor tissues and their clinical significance. Gene knockout technology using short hairpin RNA (shRNA) was used to knockdown expression of HIF-1α or Aurora A and analyze their effects on malignant phenotypes of HCC cells. The transcriptional regulation of Aurora A by HIF-1α and the possible downstream molecular signaling pathways were also determined. Results showed that hypoxia could induce the increased expression of HIF-1α and Aurora A in HCC cells. Also, shRNA-mediated HIF-1α downregulation could lead to the decreased Aurora A expression and inhibition of growth or invasion in HCC cells. Moreover, HIF-1α could transcriptionally regulate Aurora A expression by binding to hypoxia-responsive elements in the Aurora A promoter and recruiting the coactivator-p300/CBP. Additionally, shRNA-mediated Aurora A knockdown could mimic the effects of HIF-1α downregulation on phenotypes of HCC cells, and overexpression of Aurora A could partially rescue the phenotypical changes of HCC cells induced by HIF-1α downregulation. Further research indicated that activation of Akt and p38-MAPK signaling pathways mediated the downstream effects of HIF-1α and Aurora A in HCC cells under hypoxic condition. Taken together, our findings indicated that Aurora A might be a key regulator of HIF-1α-promoting malignant phenotypes of HCC by activation of Akt and p38-MAPK signaling pathways.
Collapse
Affiliation(s)
- Shi-Yun Cui
- Department of Medical Oncology; Jinling Hospital; School of Medicine; Nanjing University; Nanjing, PR China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Cheng MW, Wang BC, Weng ZQ, Zhu XW. Clinicopathological significance of Polo-like kinase 1 (PLK1) expression in human malignant glioma. Acta Histochem 2012; 114:503-9. [PMID: 22000864 DOI: 10.1016/j.acthis.2011.09.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 09/16/2011] [Accepted: 09/18/2011] [Indexed: 10/16/2022]
Abstract
Polo-like kinase 1 (PLK1), a variety of serine/threonine-protein kinase, has been reported to play important roles in malignant transformation. The purpose of this study was to investigate the clinicopathological significance of PLK1 expression in malignant glioma. A semi-quantitative RT-PCR assay was performed to detect the expression of PLK1 mRNA in 68 cases of glioma tissues and corresponding non-cancerous brain tissues. Additionally, the correlation of PLK1 mRNA expression with clinicopathological factors or prognosis of glioma patients was statistically analyzed. Multivariate analysis of prognostic factors was performed using the Cox proportional hazard model. Small interfering RNA was used to knockdown PLK1 expression in a glioma cell line and analyze the effects of PLK1 inhibition on growth, cell cycle, apoptosis and chemo- or radiosensitivity of glioma cells. Results showed that the expression of PLK1 mRNA was significantly higher in glioma tissues than in corresponding normal brain tissues. The expression of PLK1 mRNA was closely correlated with WHO grade, KPS and tumor recurrence of glioma patients (P=0.022, 0.030 and 0.041, respectively). Meanwhile, the disease-free and overall survival rates of patients with high PLK1 mRNA expression were obviously lower than those of patients with low PLK1 mRNA expression. Multivariate analysis showed that high PLK1 mRNA expression was a poor prognostic factor for glioma patients (P=0.028). The expression of PLK1 mRNA and protein was significantly down-regulated in stably transfected U251-S cells. PLK1 down-regulation could inhibit growth, induce cell arrest in G2/M phase of cell cycle and apoptosis enhancement in glioma cells. Further, PLK1 down-regulation could enhance the sensitivity of glioma cells to cisplatin or irradiation. Thus, the status of PLK1 mRNA expression might be an independent prognostic factor for glioma patients and targeting PLK1 could be a novel strategy for chemo- or radiosensitization of human malignant gliomas.
Collapse
Affiliation(s)
- Mao-Wei Cheng
- Department of Internal Neurology, Jinling Hospital, School of Medicine, Nanjing University, Jiangsu, China
| | | | | | | |
Collapse
|
16
|
Venkataraman S, Alimova I, Tello T, Harris PS, Knipstein JA, Donson AM, Foreman NK, Liu AK, Vibhakar R. Targeting Aurora Kinase A enhances radiation sensitivity of atypical teratoid rhabdoid tumor cells. J Neurooncol 2012; 107:517-26. [PMID: 22246202 DOI: 10.1007/s11060-011-0795-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 12/27/2011] [Indexed: 10/14/2022]
Abstract
Atypical teratoid/rhabdoid tumors (ATRT) are rare, highly malignant, embryonal CNS tumors with a poor prognosis. Therapy relies on highly toxic chemotherapy and radiotherapy. To improve outcomes and decrease morbidity, more targeted therapy is required. Gene expression analysis revealed elevated expression of multiple kinases in ATRT tissues. Aurora Kinase A was one of the candidate kinases. The objective of this study was to evaluate the impact of Aurora Kinase A inhibition in ATRT cell lines. Our analysis revealed that inhibition of Aurora Kinase A induces cell death in ATRT cells and the small molecule inhibitor MLN 8237 sensitizes these cells to radiation. Furthermore, inhibition of Aurora Kinase A resulted in decreased activity of pro-proliferative signaling pathways. These data indicate that inhibition of Aurora Kinase A is a promising small molecule target for ATRT therapy.
Collapse
Affiliation(s)
- Sujatha Venkataraman
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Lo Iacono M, Monica V, Saviozzi S, Ceppi P, Bracco E, Papotti M, Scagliotti GV. Aurora Kinase A expression is associated with lung cancer histological-subtypes and with tumor de-differentiation. J Transl Med 2011; 9:100. [PMID: 21718475 PMCID: PMC3148570 DOI: 10.1186/1479-5876-9-100] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 06/30/2011] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Aurora kinase A (AURKA) is a member of serine/threonine kinase family. Several kinases belonging to this family are activated in the G2/M phase of the cell cycle being involved in mitotic chromosomal segregation. AURKA overexpression is significantly associated with neoplastic transformation in several tumors and deregulated Aurora Kinases expression leads to chromosome instability, thus contributing to cancer progression. The purpose of the present study was to investigate the expression of AURKA in non small cell lung cancer (NSCLC) specimens and to correlate its mRNA or protein expression with patients' clinico-pathological features. MATERIALS AND METHODS Quantitative real-time PCR and immunohistochemistry analysis on matched cancer and corresponding normal tissues from surgically resected non-small cell lung cancers (NSCLC) have been performed aiming to explore the expression levels of AURKA gene. RESULTS AURKA expression was significantly up-modulated in tumor samples compared to matched lung tissue (p<0.01, mean log2(FC)=1.5). Moreover, AURKA was principally up-modulated in moderately and poorly differentiated lung cancers (p<0.01), as well as in squamous and adenocarcinomas compared to the non-invasive bronchioloalveolar histotype (p=0.029). No correlation with survival was observed. CONCLUSION These results indicate that in NSCLC AURKA over-expression is restricted to specific subtypes and poorly differentiated tumors.
Collapse
Affiliation(s)
- Marco Lo Iacono
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Valentina Monica
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Silvia Saviozzi
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Paolo Ceppi
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Enrico Bracco
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Mauro Papotti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giorgio V Scagliotti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
18
|
Lee S, Cimica V, Ramachandra N, Zagzag D, Kalpana GV. Aurora A is a repressed effector target of the chromatin remodeling protein INI1/hSNF5 required for rhabdoid tumor cell survival. Cancer Res 2011; 71:3225-35. [PMID: 21521802 DOI: 10.1158/0008-5472.can-10-2167] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rhabdoid tumors (RT) are aggressive pediatric malignancies with poor prognosis. INI1/hSNF5 is a component of the chromatin remodeling SWI/SNF complex and a tumor suppressor deleted in RT. Previous microarray studies indicated that reintroduction of INI1/hSNF5 into RT cells leads to repression of a high degree of mitotic genes including Aurora Kinase A (Aurora A, STK6). Here, we found that INI1/SNF5 represses Aurora A transcription in a cell-type-specific manner. INI1-mediated repression was observed in RT and normal cells but not in non-RT cell lines. Chromatin immunoprecipitation (ChIP) assay indicated that INI1/hSNF5 associates with Aurora A promoter in RT and normal cells but not in non-RT cells. Real-time PCR and immunohistochemical analyses of primary human and mouse RTs harboring mutations in INI1/hSNF5 gene indicated that Aurora A was overexpressed/derepressed in these tumor cells, confirming that INI1/hSNF5 represses Aurora A in vivo. Knockdown of Aurora A impaired cell growth, induced mitotic arrest and aberrant nuclear division leading to decreased survival, and increased cell death and caspase 3/7-mediated apoptosis in RT cells (but not in normal cells). These results indicated that Aurora A is a direct downstream target of INI1/hSNF5-mediated repression in RT cells and that loss of INI1/hSNF5 leads to aberrant overexpression of Aurora A in these tumors, which is required for their survival. We propose that a high degree of Aurora A expression may play a role in aggressive behavior of RTs and that targeting expression or activity of this gene is a novel therapeutic strategy for these tumors.
Collapse
Affiliation(s)
- Seungjae Lee
- Department of Genetics, Albert Einstein College of Medicine, New York University, New York, NY, USA
| | | | | | | | | |
Collapse
|
19
|
Akkiz H, Bayram S, Bekar A, Akgöllü E, Ozdil B. Relationship between functional polymorphism in the Aurora A gene and susceptibility of hepatocellular carcinoma. J Viral Hepat 2010; 17:668-74. [PMID: 20002563 DOI: 10.1111/j.1365-2893.2009.01225.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Aurora A is considered a potential cancer susceptibility gene owing to overexpression or amplification of Aurora A gene that causes centrosome dysfunction, chromosome instability, tumourigenic transformation and checkpoint abnormalities. Functional coding region polymorphism F31I in the Aurora A gene has recently been shown to be associated with several human cancers, but its association with hepatocellular carcinoma (HCC) has yet to be investigated. Genetic polymorphism of Aurora A was investigated in 128 confirmed subjects with HCC and 128 cancer-free control subjects matched on age, gender, smoking and alcohol consumption by using a polymerase chain reaction-restriction fragment length polymorphism assay. Allele and genotype associations of Aurora A F31I polymorphism with HCC susceptibility were observed in comparisons between the patient and control samples (respectively; P = 0.005, P = 0.012). The proportion of the genotypes containing I31 allele in patients with HCC (39.8%) was significantly higher than that in patients without HCC (22.7%) (P = 0.003). The distribution F31I genotype was significantly associated with increased risk of HCC (P = 0.003, odds ratio = 2.26, 95% confidence interval = 1.31-3.90 for FI + II genotypes vs FF genotype). Our results suggest for the first time that the Aurora A F31I polymorphism may be a genetic susceptibility factor for HCC.
Collapse
Affiliation(s)
- H Akkiz
- Department of Gastroenterology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | | | | | | | | |
Collapse
|
20
|
Barton VN, Foreman NK, Donson AM, Birks DK, Handler MH, Vibhakar R. Aurora kinase A as a rational target for therapy in glioblastoma. J Neurosurg Pediatr 2010; 6:98-105. [PMID: 20593995 DOI: 10.3171/2010.3.peds10120] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Despite advances in the knowledge of tumor biology, the outcome of glioblastoma tumors remains poor. The design of many molecularly targeted therapies in glioblastoma has focused on inhibiting molecular abnormalities present in tumor cells compared with normal tissue rather than patient outcome-associated factors. As an alternative approach, the present study identified genes associated with shorter survival as potential therapeutic targets. It was hypothesized that inhibition of a molecular target associated with poor outcome would impact glioblastoma cell proliferation. METHODS The present study correlated patient survival data with tumor gene expression profiling and gene ontology analysis. Genes associated with shorter survival were identified and one of these was selected for therapeutic targeting in an in vitro system. Glioblastoma cell growth suppression was measured by H(3)-thymidine uptake, colony formation, and flow cytometry. RESULTS The gene expression microarray and ontology analysis revealed that genes involved in mitotic processes, including AURKA, were associated with poor prognosis in glioblastoma. Inhibition of AURKA suppressed glioblastoma cell growth. Moreover, inhibition of AURKA was synergistic with radiation in glioblastoma cells at high radiation doses. CONCLUSIONS Relative expression of AURKA may be of prognostic value and warrants further investigation with larger, prospective studies. Pharmacological inhibition of AURKA is a potentially promising therapy for glioblastoma.
Collapse
Affiliation(s)
- Valerie N Barton
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado Denver, Colorado 80045, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Loh JK, Lieu AS, Chou CH, Lin FY, Wu CH, Howng SL, Chio CC, Hong YR. Differential expression of centrosomal proteins at different stages of human glioma. BMC Cancer 2010; 10:268. [PMID: 20529377 PMCID: PMC2889899 DOI: 10.1186/1471-2407-10-268] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Accepted: 06/09/2010] [Indexed: 12/31/2022] Open
Abstract
Background High-grade gliomas have poor prognosis, requiring aggressive treatment. The aim of this study is to explore mitotic and centrosomal dysregulation in gliomas, which may provide novel targets for treatment. Methods A case-control study was performed using 34 resected gliomas, which were separated into low- and high-grade groups. Normal human brain tissue was used as a control. Using immunohistochemical analysis, immunofluorescent microscopy, and RT-PCR, detection of centrins 1 and 2, γ-tubulin, hNinein, Aurora A, and Aurora B, expression was performed. Analysis of the GBM8401 glioma cell line was also undertaken to complement the in vivo studies. Results In high-grade gliomas, the cells had greater than two very brightly staining centrioles within large, atypical nuclei, and moderate-to-strong Aurora A staining. Comparing with normal human brain tissue, most of the mRNAs expression in gliomas for centrosomal structural proteins, including centrin 3, γ-tubulin, and hNinein isoforms 1, 2, 5 and 6, Aurora A and Aurora B were elevated. The significant different expression was observed between high- and low-grade glioma in both γ-tubulin and Aurora A mRNA s. In the high-grade glioma group, 78.6% of the samples had higher than normal expression of γ-tubulin mRNA, which was significantly higher than in the low-grade glioma group (18.2%, p < 0.05). Conclusions Markers for mitotic dysregulation, such as supernumerary centrosomes and altered expression of centrosome-related mRNA and proteins were more frequently detected in higher grade gliomas. Therefore, these results are clinically useful for glioma staging as well as the development of novel treatments strategies.
Collapse
Affiliation(s)
- Joon-Khim Loh
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|