Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Diagn Pathol ; 16(1): 98, 2021 Oct 27.
Article En | MEDLINE | ID: mdl-34706741

BACKGROUND: Aurora-A kinase is important for cellular proliferation and is implicated in the tumorigenesis of several malignancies, including of the ovary. Information regarding the expression patterns of Aurora-A in normal Müllerian epithelium as well as benign, borderline and malignant epithelial ovarian neoplasms is limited. METHODS: We investigated Aurora-A expression by immunohistochemistry in 15 benign, 19 borderline and 17 malignant ovarian serous tumors, and 16 benign, 8 borderline, and 2 malignant ovarian mucinous tumors. Twelve fimbriae from seven patients served as normal Müllerian epithelium controls. We also examined Aurora-A protein expression by western blot in normal fimbriae and tumor specimens. RESULTS: All normal fimbriae (n = 12) showed nuclear but not cytoplasmic Aurora-A immunoreactivity by immunohistochemistry. Benign ovarian tumors also showed strong nuclear Aurora-A immunoreactivity. Forty-eight percent (13/27) of borderline tumors demonstrated nuclear Aurora-A immunoreactivity, while the remainder (52%, 14/27) lacked Aurora-A staining. Nuclear Aurora-A immunoreactivity was absent in all malignant serous tumors, however, 47% (8/17) demonstrated perinuclear cytoplasmic staining. These results were statistically significant when tumor class (benign/borderline/malignant) was compared to immunoreactivity localization or intensity (Fisher Exact Test, p < 0.01). Western blot analysis confirmed the greater nuclear Aurora-A expression in control Müllerian epithelium compared to borderline and malignant tumors. CONCLUSION: Aurora-A kinase is differentially expressed across normal Müllerian epithelium, benign and borderline serous and mucinous ovarian epithelial neoplasms and malignant serous ovarian tumors., with nuclear expression of unphosphorylated Aurora-A being present in normal and benign neoplastic epithelium, and lost in malignant serous neoplasms. Further studies of the possible biological and clinical implications of the loss of nuclear Aurora-A expression in ovarian tumors, and its role in ovarian carcinogenesis are warranted.


Aurora Kinase A/biosynthesis , Carcinoma, Ovarian Epithelial/enzymology , Cystadenocarcinoma, Mucinous/enzymology , Cystadenocarcinoma, Serous/enzymology , Ovary/enzymology , Carcinoma, Ovarian Epithelial/pathology , Cell Nucleus/enzymology , Cystadenocarcinoma, Mucinous/pathology , Cystadenocarcinoma, Serous/pathology , Cytoplasm/enzymology , Epithelium/enzymology , Female , Humans
2.
Reprod Biol Endocrinol ; 19(1): 162, 2021 Oct 29.
Article En | MEDLINE | ID: mdl-34715887

BACKGROUND: Decidualization is essential to the successful pregnancy in mice. The molecular mechanisms and effects of Aurora kinase A (Aurora A) remain poorly understood during pregnancy. This study is the first to investigate the expression and role of Aurora A during mouse decidualization. METHODS: Quantitative real time polymerase chain reaction, western blotting and in situ hybridization were used to determine the expression of Aurora A in mouse uteri. Aurora A activity was inhibited by Aurora A inhibitor to explore the role of Aurora A on decidualization via regulating the Aurora A/Stat3/Plk1/Cdk1 signaling pathway. RESULTS: Aurora A was strongly expressed at implantation sites compared with inter-implantation sites. Furthermore, Aurora A was also significantly increased in oil-induced deciduoma compared with control. Both Aurora A mRNA and protein were significantly increased under in vitro decidualization. Under in vitro decidualization, Prl8a2, a marker of mouse decidualization, was significantly decreased by TC-S 7010, an Aurora A inhibitor. Additionally, Prl8a2 was reduced by Stat3 inhibitor, Plk1 inhibitor and Cdk1 inhibitor, respectively. Moreover, the protein levels of p-Stat3, p-Plk1 and p-Cdk1 were suppressed by TC-S 7010. The protein levels of p-Stat3, p-Plk1 and p-Cdk1 were also suppressed by S3I-201, a Stat3 inhibitor). SBE 13 HCl (Plk1 inhibitor) could reduce the protein levels of p-Plk1 and p-Cdk1. Collectively, Aurora A could regulate Stat3/Plk1/Cdk1 signaling pathway. CONCLUSION: Our study shows that Aurora A is expressed in decidual cells and should be important for mouse decidualization. Aurora A/Stat3/Plk1/Cdk1 signaling pathway may be involved in mouse decidualization.


Aurora Kinase A/biosynthesis , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Decidua/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Animals , Aurora Kinase A/antagonists & inhibitors , CDC2 Protein Kinase/antagonists & inhibitors , Cell Cycle Proteins/antagonists & inhibitors , Cells, Cultured , Decidua/drug effects , Enzyme Inhibitors/pharmacology , Female , Mice , Pregnancy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Polo-Like Kinase 1
3.
Life Sci ; 271: 119148, 2021 Apr 15.
Article En | MEDLINE | ID: mdl-33545203

AIMS: Abnormally amplified expression of AURKA (aurora kinase A) is closely related to chemo-resistance in human colorectal cancer, lung cancer and leukemia. However, the biological role of AURKA in response to radio-sensitivity in human colorectal cancer is still unknown. Therefore, we evaluated the radio-sensitize ability of perturbation AURKA in human colorectal cancer. MAIN METHODS: The knockdown effect of shAURKA was determined by western blot and qRT-PCR, respectively. Cell growth was determined by CCK-8 and clonogenic assay. Cell migration and metastasis was measured by wound healing assay and transwell invasive assay, respectively. Cell cycle and apoptosis was analyzed by flow cytometry. The alteration of down-stream targets was determined by western blot analysis. KEY FINDINGS: We observed that high-level of AURKA expression is associated with poor prognosis in CRC patients receiving radiotherapy. Knockdown of AURKA significantly sensitizes the efficacy of radiation on the proliferation of HCT116 and HT-29 cells. The combination of AURKA inhibition and radiation could effectively suppress the ability of cell migration and metastasis, but also synergistically induce cellular apoptosis and arrest cell cycle at G2/M phase. Further studies demonstrated that knockdown AURKA markedly enhanced the efficacy of radiation through elevated PARP cleavage and induced AURKA-mediated pro-apoptosis factor BIM. Meanwhile, knockdown of AURKA in combination with radiation synergistically suppressed the regulator in blockage of G2/M phase, CDK2. SIGNIFICANCE: Taken together, our results provide the evidence that targeted inhibition of AURKA could be a promising strategy for enhancing the efficacy of radiation for the treatment of human colorectal cancer.


Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/radiotherapy , Gene Expression Regulation, Neoplastic , Aurora Kinase A/biosynthesis , Cell Proliferation/radiation effects , Colorectal Neoplasms/metabolism , Gene Knockdown Techniques/methods , HCT116 Cells , HT29 Cells , Humans , Treatment Outcome
4.
PLoS One ; 15(5): e0233717, 2020.
Article En | MEDLINE | ID: mdl-32469983

Metastasis is known as a key step in cancer recurrence and could be stimulated by multiple factors. Calumenin (CALU) is one of these factors which has a direct impact on cancer metastasis and yet, its underlined mechanisms have not been completely elucidated. The current study was aimed to identify CALU co-expressed genes, their signaling pathways, and expression status within the human cancers. To this point, CALU associated genes were visualized using the Cytoscape plugin BisoGenet and annotated with the Enrichr web-based application. The list of CALU related diseases was retrieved using the DisGenNet, and cancer datasets were downloaded from The Cancer Genome Atlas (TCGA) and analyzed with the Cufflink software. ROC curve analysis was used to estimate the diagnostic accuracy of DEGs in each cancer, and the Kaplan-Meier survival analysis was performed to plot the overall survival of patients. The protein level of the signature biomarkers was measured in 40 biopsy specimens and matched adjacent normal tissues collected from CRC and lung cancer patients. Analysis of CALU co-expressed genes network in TCGA datasets indicated that the network is markedly altered in human colon (COAD) and lung (LUAD) cancers. Diagnostic accuracy estimation of differentially expressed genes showed that a gene panel consisted of CALU, AURKA, and MCM2 was able to successfully distinguish cancer tumors from healthy samples. Cancer cases with abnormal expression of the signature genes had a significantly lower survival rate than other patients. Additionally, comparison of CALU, AURKA, and MCM2 proteins between healthy samples, early and advanced tumors showed that the level of these proteins was increased through normal-carcinoma transition in both types of cancers. These data indicate that the interactions between CALU, AURKA, and MCM2 has a pivotal role in cancer development, and thereby needs to be explored in the future.


Aurora Kinase A , Calcium-Binding Proteins , Colonic Neoplasms , Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Minichromosome Maintenance Complex Component 2 , Aurora Kinase A/biosynthesis , Aurora Kinase A/genetics , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Colonic Neoplasms/diagnosis , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Disease-Free Survival , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Minichromosome Maintenance Complex Component 2/biosynthesis , Minichromosome Maintenance Complex Component 2/genetics , Neoplasm Metastasis , Survival Rate
5.
Am J Physiol Cell Physiol ; 317(3): C600-C612, 2019 09 01.
Article En | MEDLINE | ID: mdl-31314582

Mitosis is a complicated process by which eukaryotic cells segregate duplicated genomes into two daughter cells. To achieve the goal, numerous regulators have been revealed to control mitosis. The oncogenic Aurora-A is a versatile kinase responsible for the regulation of mitosis including chromosome condensation, spindle assembly, and centrosome maturation through phosphorylating a range of substrates. However, overexpression of Aurora-A bypasses cytokinesis, thereby generating multiple nuclei by unknown the mechanisms. To explore the underlying mechanisms, we found that SLAN, a potential tumor suppressor, served as a substrate of Aurora-A and knockdown of SLAN induced immature cytokinesis. Aurora-A phosphorylates SLAN at T573 under the help of the scaffold protein 14-3-3η. The SLAN phosphorylation-mimicking mutants T573D or T573E, in contrast to the phosphorylation-deficiency mutant T573A, induced higher level of multinucleated cells, and the endogenous SLAN p573 resided at spindle midzone and midbody with the help of the microtubule motor MKLP1. The Aurora-A- or SLAN-induced multiple nuclei was prevented by the knockdown of 14-3-3η or Aurora-A respectively, thereby revealing a 14-3-3η/Aurora-A/SLAN cascade negatively controlling cytokinesis. Intriguingly, SLAN T573D or T573E inactivated and T573A activated the key cytokinesis regulator RhoA. RhoA interacted with SLAN np573, i.e., the nonphosphorylated form of SLAN at T573, which localized to the spindle midzone dictated by RhoA and ECT2. Therefore, we report here that SLAN mediates the Aurora-A-triggered cytokinesis bypass and SLAN plays dual roles in that process depending on its phosphorylation status.


Aurora Kinase A/biosynthesis , Cytokinesis/physiology , Gene Expression Regulation, Enzymologic , Tumor Suppressor Proteins/metabolism , Aurora Kinase A/genetics , HEK293 Cells , Humans , Phosphorylation/physiology
6.
Eur Rev Med Pharmacol Sci ; 23(10): 4275-4284, 2019 May.
Article En | MEDLINE | ID: mdl-31173299

OBJECTIVE: Increasing evidence suggested that dysregulated miR-154 in several tumor tissues is involved in the clinical progress of cancers patients. The objective of this study was to explore the expression pattern of miR-154 and its potential effects in human melanoma. PATIENTS AND METHODS: Microarray data from GEO datasets were analyzed to identify differentially expressed miRNAs. Real Time-Polymerase Chain Reaction (RT-PCR) was performed to determine the expressions of miR-154 in melanoma cell lines and tumor tissues. The associations between miR-154 levels and clinical progress were studied using a series of statistical methods. Cell viability, invasion, migration, and apoptosis were detected by Cell Counting Kit-8 (CCK-8) assays, transwell assay, wound healing assays, and flow cytometry, respectively. TargetScan system was used to identify the target genes of miR-154 and Luciferase activity analysis was carried out to demonstrate the possible target. RESULTS: The expression levels of miR-154 were distinctly lower in tumor samples and melanoma cell lines than in normal controls (p < 0.01). The up-regulation of miR-154 in melanoma tissues was associated with advanced tumor stage (p = 0.028), ulceration (p = 0.046), and shorter overall survival (p = 0.0035). Moreover, the multivariate analysis suggested a decreased expression of miR-154 is an independent predictor of overall survival rates in melanoma patients. Functional observation showed that up-regulation of miR-154 suppressed the capability of proliferation, invasion, and migration, promoting apoptosis in melanoma cell lines. Bioinformatics analysis predicted AURKA (aurora kinase A) as a target of miR-154, which was confirmed using the luciferase activity assays. Besides, miR-154 overexpression rescued the suppressive effect of AURKA-mediated melanoma on cell proliferation, colony formation, and metastasis. CONCLUSIONS: These results revealed that miR-154 has clinical implications for targeted therapy of melanoma patients and indicated that miR-154 could represent a novel biomarker in predicting the clinical outcome for melanoma.


Aurora Kinase A/genetics , Melanoma/genetics , MicroRNAs/genetics , Adult , Aurora Kinase A/biosynthesis , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Cell Count , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Melanoma/pathology , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Prognosis , Survival Analysis , Tumor Stem Cell Assay , Up-Regulation
7.
Cancer Res ; 79(5): 928-940, 2019 03 01.
Article En | MEDLINE | ID: mdl-30498084

The oncogenic phosphatase PRL-3 is highly expressed in metastatic colorectal cancer but not in nonmetastatic colorectal cancer or noncolorectal cancer metastatic cancers. Although the proinvasive capacity of PRL-3 has been validated in multiple types of cancer, its impact on colorectal cancer progression and the underlying mechanisms remain poorly understood. Here, we report that overexpressed PRL-3 stimulates G2-M arrest, chromosomal instability (CIN), self-renewal, and growth of colorectal cancer cells in xenograft models, while colorectal cancer cell proliferation is decreased. PRL-3-induced G2-M arrest was associated with decreased expression of Aurora kinase A (AURKA). PRL-3-promoted slow proliferation, CIN, self-renewal, and growth in xenografts were counteracted by ectopic expression of AURKA. Conversely, knockdown of PRL-3 resulted in low proliferation, S-phase arrest, impaired self-renewal, increased apoptosis, and diminished xenograft growth independently of AURKA. Analysis of colorectal cancer specimens showed that expression of PRL-3 was associated with high status of CIN and poor prognosis, which were antagonized by expression of AURKA. PRL-3 enhanced AURKA ubiquitination and degradation in a phosphatase-dependent fashion. PRL-3 interacted with AURKA and FZR1, a regulatory component of the APC/CFZR1 complex. Destabilization of AURKA by PRL-3 required PRL-3-mediated dephosphorylation of FZR1 and assembly of the APC/CFZR1 complex. Our study suggests that PRL-3-regulated colorectal cancer progression is collectively determined by distinct malignant phenotypes and further reveals PRL-3 as an essential regulator of APC/CFZR1 in controlling the stability of AURKA. SIGNIFICANCE: Dephosphorylation of FZR1 by PRL-3 facilitates the activity of APC/CFZR1 by destabilizing AURKA, thus influencing aggressive characteristics and overall progression of colorectal cancer.


Aurora Kinase A/metabolism , Cdh1 Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Aurora Kinase A/biosynthesis , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Female , G2 Phase Cell Cycle Checkpoints , HCT116 Cells , HT29 Cells , HeLa Cells , Heterografts , Humans , M Phase Cell Cycle Checkpoints , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/biosynthesis , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Tyrosine Phosphatases/biosynthesis , Ubiquitin/metabolism , Ubiquitination
8.
Int J Cancer ; 142(1): 92-108, 2018 01 01.
Article En | MEDLINE | ID: mdl-28884479

In this study, we firstly showed that p53 transcriptionally represses Aurora-A gene expression through directly binding to its promoter. DNA affinity precipitation assay and chromatin immunoprecipitation assay indicated that p53 physically bound to the Aurora-A promoter. Moreover, the in vitro and in vivo assays showed that p53 directly bound to the Aurora-A promoter together with histone deacetylase 1 (HDAC1) and mSin3a as corepressors. Furthermore, we identified that the nucleotides -360 to -354 (CCTGCCC), upstream of the Aurora-A transcriptional start site, was responsible for the p53-mediated repression. Mutation within this site disrupted its interaction with p53, mSin3a and HDAC1, as well as attenuated the repressive effect of p53 on Aurora-A promoter activity. Treatment with trichostatin A (TSA), a HDAC1 inhibitor, disrupted the interaction of p53-HDAC1-mSin3a complex with the nucleotides -365∼-345 region, and enhanced the Aurora-A promoter activity and gene expression. Additionally, knockdown of p53 or mSin3a also drastically blocked the formation of p53-HDAC1-mSin3a repressive complex onto this promoter region and elevated the Aurora-A promoter activity and gene expression. Moreover, the p53-HDAC1-mSin3a repressive complex also involved in the inhibition of Aurora-A gene expression upon cisplatin treatment. Finally, the clinical investigation showed that Aurora-A and p53 exhibited an inverse correlation in both the expression level and prognostic status, and the low p53/high Aurora-A showed the poorest prognosis of NSCLC patients. Our findings showed novel regulatory mechanisms of p53 in regulating Aurora-A gene expression in NSCLC cells.


Adenocarcinoma/genetics , Aurora Kinase A/biosynthesis , Gene Expression Regulation, Neoplastic/genetics , Lung Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Adenocarcinoma/mortality , Aurora Kinase A/genetics , Cell Line, Tumor , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics
9.
Exp Neurol ; 299(Pt B): 299-307, 2018 01.
Article En | MEDLINE | ID: mdl-28610844

Meningiomas frequently display activation of the PI3K/AKT/mTOR pathway, leading to elevated levels of phospho-eukaryotic translation initiation factor 4E binding proteins, which enhances protein synthesis; however, it is not known whether inhibition of protein translation is an effective treatment option for meningiomas. We found that human meningiomas expressed high levels of the three components of the eukaryotic initiation factor 4F (eIF4F) translation initiation complex, eIF4A, eIF4E, and eIF4G. The expression of eIF4A and eIF4E was important in sustaining the growth of NF2-deficient benign meningioma Ben-Men-1 cells, as shRNA-mediated knockdown of these proteins strongly reduced cell proliferation. Among a series of 23 natural compounds evaluated, silvestrol, which inhibits eIF4A, was identified as being the most growth inhibitory in both primary meningioma and Ben-Men-1 cells. Silvestrol treatment of meningioma cells prominently induced G2/M arrest. Consistently, silvestrol significantly decreased the amounts of cyclins D1, E1, A, and B, PCNA, and Aurora A. In addition, total and phosphorylated AKT, ERK, and FAK, which have been shown to be important drivers for meningioma cell proliferation, were markedly lower in silvestrol-treated Ben-Men-1 cells. Our findings suggest that inhibiting protein translation could be a potential treatment for meningiomas.


Antineoplastic Agents/pharmacology , Eukaryotic Initiation Factor-4A/biosynthesis , Eukaryotic Initiation Factor-4E/biosynthesis , Eukaryotic Initiation Factor-4G/biosynthesis , Meningeal Neoplasms/drug therapy , Meningioma/drug therapy , Neoplasm Proteins/biosynthesis , Peptide Chain Initiation, Translational/drug effects , Triterpenes/pharmacology , Antineoplastic Agents/therapeutic use , Aurora Kinase A/biosynthesis , Aurora Kinase A/genetics , Cyclins/biosynthesis , Cyclins/genetics , Drug Screening Assays, Antitumor , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4E/genetics , Eukaryotic Initiation Factor-4G/genetics , Female , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Male , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Meningioma/genetics , Meningioma/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Proliferating Cell Nuclear Antigen/biosynthesis , Proliferating Cell Nuclear Antigen/genetics , RNA, Small Interfering/pharmacology , Triterpenes/therapeutic use , Tumor Cells, Cultured
11.
Int J Gynecol Cancer ; 27(8): 1666-1674, 2017 10.
Article En | MEDLINE | ID: mdl-28786875

OBJECTIVE: This study aims to clarify the incidence of Aurora kinase A (Aurora-A) protein expression and its correlation with clinical parameters in ovarian clear cell carcinoma (OCCC) tumor tissues. In addition, we assessed the efficacy of ENMD-2076, a novel selective Aurora-A inhibitor, in combination with chemotherapeutic agents for the treatment of OCCC. METHODS/MATERIALS: Aurora-A protein expression was determined by immunohistochemical staining of OCCC specimens from 56 patients to evaluate its correlation with clinical outcomes in OCCC. In the in vitro study, 6 OCCC cell lines were exposed to ENMD-2076 in combination with cisplatin, SN38, doxorubicin, or paclitaxel, and cell proliferation, cell cycle distribution, and apoptosis were assessed. RESULTS: The 5-year survival rates of International Federation of Gynecology and Obstetrics stages IC3 to IV patients with intermediate or strong Aurora-A expression were significantly lower than those of patients with negative or weak Aurora-A expression. Increased Aurora-A expression was associated with significantly worse overall survival of International Federation of Gynecology and Obstetrics stages IC3 to IV patients (21% vs 77%). Multivariate analysis revealed that Aurora-A expression was an independent prognostic factor for stages IC3 to IV OCCC patients. Furthermore, synergistic effects were observed with ENMD-2076 in combination with cisplatin or SN-38 in 4 of the 6 tested cell lines. ENMD-2076 dramatically enhanced apoptosis and cell cycle arrest at the G2/M phase induced by cisplatin. CONCLUSIONS: Aurora-A is a promising biomarker that is predictive of patient outcomes and a potential target for OCCC. The results suggested that chemotherapy, including ENMD-2076 in combination with cisplatin, is a potential treatment modality for patients with OCCC.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aurora Kinase A/antagonists & inhibitors , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/enzymology , Adult , Aged , Apoptosis/drug effects , Aurora Kinase A/biosynthesis , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/biosynthesis , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Cisplatin/administration & dosage , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Synergism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Immunohistochemistry , Irinotecan , Ki-67 Antigen/biosynthesis , M Phase Cell Cycle Checkpoints/drug effects , Middle Aged , Ovarian Neoplasms/enzymology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Prognosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Tissue Array Analysis
12.
Int J Oncol ; 50(5): 1868-1878, 2017 May.
Article En | MEDLINE | ID: mdl-28393219

Multiple lines of evidence indicate that aberrant activation of Hedgehog (Hh) signaling plays an important role in tumorigenesis in human glioma. However, the underlying molecular mechanism and crucial downstream targets of glioma-associated oncogene (Gli), a primary transcriptional regulator of Hh signaling, are not fully understood. Here, we report the identification of miR-124 as a novel downstream target of the transcriptional factor Gli2, which is important for proliferation and tumor growth in human glioma cells. Blockade of Hh signaling leads to a remarkable increase in miR-124 expression in glioma cells, whereas overexpression of Gli2 suppresses miR-124 expression by increasing the direct binding of Gli2 to the upstream region of the transcriptional start site for miR-124. Furthermore, we found that miR-124 potentially interacts with the 3'-UTR region of AURKA. Overexpression of miR-124 significantly decreased the expression of AURKA in glioma cells. In contrast, the loss of miR-124 led to the increased expression of AURKA mRNA and protein. In addition, cell proliferation and colony formation ability were significantly decreased following Gli2 knockdown in human glioma cells, while transfection with a miR-124 inhibitor rescued the proliferative ability of cells. These results demonstrate that miR-124 is an important downstream target gene of Hh signaling, and the Gli2/miR-124/AURKA axis is essential for the proliferation and growth of human glioma cells.


Aurora Kinase A/biosynthesis , Glioma/genetics , Hedgehog Proteins/genetics , Kruppel-Like Transcription Factors/biosynthesis , MicroRNAs/biosynthesis , Nuclear Proteins/biosynthesis , Aurora Kinase A/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Glioma/pathology , Hedgehog Proteins/antagonists & inhibitors , Humans , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , Nuclear Proteins/genetics , Signal Transduction/drug effects , Transcription Factors/genetics , Zinc Finger Protein Gli2
13.
BMC Cancer ; 17(1): 277, 2017 04 18.
Article En | MEDLINE | ID: mdl-28420331

BACKGROUND: Arsenic is a widely distributed metalloid compound that has biphasic effects on cultured cells. In large doses, arsenic can be toxic enough to trigger cell death. In smaller amounts, non-toxic doses may promote cell proliferation and induces carcinogenesis. Aberration of chromosome is frequently detected in epithelial cells and lymphocytes of individuals from arsenic contaminated areas. Overexpression of Aurora-A, a mitotic kinase, results in chromosomal instability and cell transformation. We have reported that low concentration (≦1 µM) of arsenic treatment increases Aurora-A expression in immortalized bladder urothelial E7 cells. However, how arsenic induces carcinogenesis through Aurora-A activation remaining unclear. METHODS: Bromodeoxyuridine (BrdU) staining, MTT assay, and flow cytometry assay were conducted to determine cell proliferation. Messenger RNA and protein expression levels of Aurora-A were detected by reverse transcriptional-PCR and Western blotting, respectively. Centrosome of cells was observed by immunofluorescent staining. The transcription factor of Aurora-A was investigated by promoter activity, chromosome immunoprecipitation (ChIP), and small interfering RNA (shRNA) assays. Mouse model was utilized to confirm the relationship between arsenic and Aurora-A. RESULTS: We reveal that low dosage of arsenic treatment increased cell proliferation is associated with accumulated cell population at S phase. We also detected increased Aurora-A expression at mRNA and protein levels in immortalized bladder urothelial E7 cells exposed to low doses of arsenic. Arsenic-treated cells displayed increased multiple centrosome which is resulted from overexpressed Aurora-A. Furthermore, the transcription factor, E2F1, is responsible for Aurora-A overexpression after arsenic treatment. We further disclosed that Aurora-A expression and cell proliferation were increased in bladder and uterus tissues of the BALB/c mice after long-term arsenic (1 mg/L) exposure for 2 months. CONCLUSION: We reveal that low dose of arsenic induced cell proliferation is through Aurora-A overexpression, which is transcriptionally regulated by E2F1 both in vitro and in vivo. Our findings disclose a new possibility that arsenic at low concentration activates Aurora-A to induce carcinogenesis.


Arsenic/toxicity , Aurora Kinase A/biosynthesis , Carcinoma, Transitional Cell/enzymology , E2F1 Transcription Factor/metabolism , Urinary Bladder Neoplasms/enzymology , Animals , Blotting, Western , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction
14.
Int J Oncol ; 50(2): 365-372, 2017 Feb.
Article En | MEDLINE | ID: mdl-28101582

The growth of a tumor depends to a certain extent on an increase in mitotic events. Key steps during mitosis are the regulated assembly of the spindle apparatus and the separation of the sister chromatids. The microtubule-associated protein Aurora kinase A phosphorylates DLGAP5 in order to correctly segregate the chromatids. Its activity and recruitment to the spindle apparatus is regulated by TPX2. KIF11 and CKAP5 control the correct arrangement of the microtubules and prevent their degradation. In the present study, we investigated the role of these five molecules in non-small cell lung cancer (NSCLC). We analyzed the expression of the five genes in a large cohort of NSCLC patients (n=362) by quantitative real-time PCR. Each of the genes was highly overexpressed in the tumor tissues compared to corresponding normal lung tissue. The correlation of the expression of the individual genes depended on the histology. An increased expression of AURKA, DLGAP5, TPX2, KIF11 and CKAP5 was associated with poor overall survival (P=0.001-0.065). AURKA was a significant prognostic marker using multivariate analyses (P=0.006). Immunofluorescence studies demonstrated that the five mitosis-associated proteins co-localized with the spindle apparatus during cell division. Taken together, our data demonstrate that the expression of the mitosis-associated genes AURKA, DLGAP5, TPX2, KIF11 and CKAP5 is associated with the prognosis of NSCLC patients.


Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Aurora Kinase A/biosynthesis , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Cell Cycle Proteins/biosynthesis , Female , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Kinesins/biosynthesis , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Microtubule-Associated Proteins/biosynthesis , Middle Aged , Mitosis/genetics , Neoplasm Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Prognosis , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Transcriptome
15.
Carcinogenesis ; 37(12): 1180-1189, 2016 Dec.
Article En | MEDLINE | ID: mdl-27624071

Recent data from The Cancer Genome Atlas analysis have revealed that Aurora kinase A (AURKA) amplification and overexpression characterize a distinct subset of human tumors across multiple cancer types. Although elevated expression of AURKA has been shown to induce oncogenic phenotypes in cells in vitro, findings from transgenic mouse models of Aurora-A overexpression in mammary glands have been distinct depending on the models generated. In the present study, we report that prolonged overexpression of AURKA transgene in mammary epithelium driven by ovine ß-lactoglobulin promoter, activated through multiple pregnancy and lactation cycles, results in the development of mammary adenocarcinomas with alterations in cancer-relevant genes and epithelial-to-mesenchymal transition. The tumor incidence was 38.9% (7/18) in Aurora-A transgenic mice at 16 months of age following 4-5 pregnancy cycles. Aurora-A overexpression in the tumor tissues accompanied activation of Akt, elevation of Cyclin D1, Tpx2 and Plk1 along with downregulation of ERα and p53 proteins, albeit at varying levels. Microarray comparative genomic hybridization (CGH) analyses of transgenic mouse mammary adenocarcinomas revealed copy gain of Glp1r and losses of Ercc5, Pten and Tcf7l2 loci. Review of human breast tumor transcriptomic data sets showed association of these genes at varying levels with Aurora-A gain of function alterations. Whole exome sequencing of the mouse tumors also identified gene mutations detected in Aurora-A overexpressing human breast cancers. Our findings demonstrate that prolonged overexpression of Aurora-A can be a driver somatic genetic event in mammary adenocarcinomas associated with deregulated tumor-relevant pathways in the Aurora-A subset of human breast cancer.


Adenocarcinoma/genetics , Aurora Kinase A/biosynthesis , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/genetics , Mammary Neoplasms, Animal/genetics , Adenocarcinoma/pathology , Animals , Aurora Kinase A/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelium/metabolism , Epithelium/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Mice , Neoplasm Proteins/biosynthesis , Pregnancy
16.
Anticancer Res ; 36(8): 3883-7, 2016 Aug.
Article En | MEDLINE | ID: mdl-27466490

BACKGROUND/AIM: Rhabdoid tumors (RT) are aggressive pediatric tumors, which show poor prognosis despite use of multimodal intensive therapy. In these tumors, several different oncogenic pathways and epigenetic regulators (like CDK4/6-cyclinD-Rb-signaling, EZH2, histone deacetylases) are contemporaneously deregulated as a consequence of biallelic SMARCB1/SNF5/INI1 alterations. Since these tumors are highly resistant to current therapies, alternative treatment strategies are urgently required. MATERIALS AND METHODS: In this study, we evaluated cytotoxic effects (by MTT tests) of small molecular compounds, which specifically target these deregulated pathways, using either single-drug or combined approaches. Half-maximal inhibitory concentration (IC50) and combined index (CI) were calculated. RESULTS: All target-directed inhibitors blocked cell growth of three different rhabdoid tumor cell lines in vitro. Several combinations of those target-specific drugs synergistically inhibited cell proliferation of rhabdoid tumors. CONCLUSION: Supporting earlier reports, combined target-directed approaches are a promising tool for the therapy of malignant rhabdoid tumors.


Aurora Kinase A/biosynthesis , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Histone Deacetylases/biosynthesis , Rhabdoid Tumor/drug therapy , SMARCB1 Protein/genetics , Aurora Kinase A/antagonists & inhibitors , Bortezomib/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Combined Modality Therapy , Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylases/drug effects , Humans , Molecular Targeted Therapy , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , SMARCB1 Protein/antagonists & inhibitors , SMARCB1 Protein/biosynthesis , Small Molecule Libraries/administration & dosage
17.
Int J Oncol ; 49(2): 823-37, 2016 Aug.
Article En | MEDLINE | ID: mdl-27278006

We have recently shown that mRNA and protein of PHLDA1 (pleckstrin-homology-like domain family A, member  1) were by far the most upregulated molecules upon treatment of IMR-32 cells with the anti-GD2 ganglioside monoclonal antibody 14G2a. Hence, we decided to study functions of PHLDA1 using human neuroblastoma IMR-32 cells as a model. Here, we show that constitutive expression of mRNA and protein of the PHLDA1 gene in IMR-32 cells was inversely correlated with transcript of the AURKA gene and Aurora A oncoprotein. Next, we silenced PHLDA1 expression in IMR-32 cells using an shRNA interference method. We report that IMR-32 cells with stable downregulation of PHLDA1 showed enhanced cellular ATP levels and an increase in mitochondrial membrane potential, as compared to control and non-transduced cells. We demonstrated that downregulation of PHLDA1 leads to a significant increase in expression of Aurora A and TRKB that are markers of poor prognosis in neuroblastoma. Also, we measured an increase in Aurora A and Akt kinases phosphorylation in the cells. Most importantly, PHLDA1-silenced cells were less susceptible to apoptosis than control cells, as shown by the lower expression of cleaved caspase-3 and PARP as well as a decreased activity of caspase-3 and -7. Our study negatively correlates expression of PHLDA1 and Aurora A in IMR-32 cells and sheds new light on functions of PHLDA1 in the neuroblastoma tumor cells, suggesting its role as a pro-apoptotic protein. Additionally, our results show possible links of the protein to regulation of features of mitochondria and formation of autophagosomes.


Aurora Kinase A/biosynthesis , Membrane Glycoproteins/biosynthesis , Neuroblastoma/genetics , Protein-Tyrosine Kinases/biosynthesis , Transcription Factors/genetics , Adenosine Triphosphate/biosynthesis , Apoptosis/genetics , Aurora Kinase A/genetics , Autophagy/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/genetics , Membrane Potential, Mitochondrial/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neuroblastoma/pathology , Protein-Tyrosine Kinases/genetics , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , Receptor, trkB , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis
18.
Oncotarget ; 7(29): 45803-45818, 2016 Jul 19.
Article En | MEDLINE | ID: mdl-27329586

Development of colorectal cancer (CRC) involves sequential transformation of normal mucosal tissues into benign adenomas and then adenomas into malignant tumors. The identification of genes crucial for malignant transformation in colorectal adenomas (CRAs) has been based primarily on cross-sectional observations. In this study, we identified relevant genes using autologous samples. By performing genome-wide SNP genotyping and RNA sequencing analysis of adenocarcinomas, adenomatous polyps, and non-neoplastic colon tissues (referred as tri-part samples) from individual patients, we identified 68 genes with differential copy number alterations and progressively dysregulated expression. Aurora A, SKA3, and DSN1 protein levels were sequentially up-regulated in the samples, and this overexpression was associated with chromosome instability (CIN). Knockdown of SKA3 in CRC cells dramatically reduced cell growth rates and increased apoptosis. Depletion of SKA3 or DSN1 induced G2/M arrest and decreased migration, invasion, and anchorage-independent growth. AURKA and DSN1 are thus critical for chromosome 20q amplification-associated malignant transformation in CRA. Moreover, SKA3 at chromosome 13q was identified as a novel gene involved in promoting malignant transformation. Evaluating the expression of these genes may help identify patients with progressive adenomas, helping to improve treatment.


Adenocarcinoma/pathology , Adenoma/pathology , Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/pathology , Adenocarcinoma/genetics , Adenoma/genetics , Adult , Aged , Area Under Curve , Aurora Kinase A/biosynthesis , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/biosynthesis , Colorectal Neoplasms/genetics , Disease Progression , Female , Gene Expression Profiling , Humans , Male , Microtubule-Associated Proteins/biosynthesis , Middle Aged , ROC Curve , Transcriptome , Up-Regulation
19.
Carcinogenesis ; 37(7): 701-711, 2016 07.
Article En | MEDLINE | ID: mdl-27207661

Neuroendocrine prostate cancer (NEPC) has a poor prognosis, with a median survival of less than 1 year after diagnosis. Following androgen deprivation therapy, prostate adenocarcinoma cells have been observed to develop an androgen receptor-negative, terminally differentiated and indolent neuroendocrine-like phenotype. However, several molecular events, including interleukin 6 (IL-6) stimulation, in the prostate microenvironment result in the appearance of aggressive, highly proliferative castrate-resistant NEPC. In this study, we examined the mechanistic effects of a natural prenylflavonoid, icaritin (ICT), on neuroendocrine differentiation in IL-6-induced LNCaP cells and NEPC development in the male transgenic adenocarcinoma of the mouse prostate (TRAMP) model. TRAMP mice received daily intraperitoneal injection of ICT or vehicle. ICT induced apoptosis in prostate tumor, suppressed NEPC development and, accordingly, improved overall survival in TRAMP mice. Expression of neuroendocrine markers (synaptophysin) and androgen receptor in TRAMP mice and neuroendocrine-like LNCaP cells were inhibited by ICT. Suppression of neuroendocrine and NEPC development by ICT was associated with dose-dependent inhibitory effects on abnormally elevated IL-6/STAT3 and Aurora kinase A in vitro and in vivo Since ICT demonstrated favorable pharmacokinetic and safety profiles with marked enrichment in prostate tissues, our study provides evidence for the development of prenylflavonoid as a multimodal therapeutic agent against NEPC.


Aurora Kinase A/biosynthesis , Carcinoma, Neuroendocrine/drug therapy , Flavonoids/administration & dosage , Interleukin-6/biosynthesis , Prostatic Neoplasms/drug therapy , STAT3 Transcription Factor/biosynthesis , Animals , Aurora Kinase A/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Cell Differentiation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/genetics , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , STAT3 Transcription Factor/genetics , Signal Transduction , Synaptophysin/biosynthesis , Synaptophysin/genetics , Tumor Microenvironment/drug effects
20.
Exp Cell Res ; 340(1): 91-101, 2016 Jan 01.
Article En | MEDLINE | ID: mdl-26658517

Soft tissue sarcomas are relatively rare, unusual, anatomically diverse group of malignancies. According to the recent literature and medical bulletins, tumor growth and aggressiveness immensely relies on its anatomical locations. However, it is unclear whether the cranio-caudal anatomical axis of the mammalian body can influence sarcoma development and the underlying molecular mechanisms are not yet deciphered. Here, we investigated the growth pattern of solid sarcoma implanted into the murine cranial and caudal anatomical locations and tried to explore the location specific expression pattern of crucial mammalian mitotic regulators such as Aurora kinase A, Histone H3 and c-Myc in the cranio-caudally originated solid tumors. In addition, the influence of local tumor microenvironment on regional sarcoma growth was also taken into consideration. We found that solid sarcoma developed differentially when implanted into two different anatomical locations and most notably, enhanced tumor growth was observed in case of cranially implanted sarcoma than the caudal sarcoma. Interestingly, Aurora kinase A and c-Myc expression and histone H3 phosphorylation level were comparatively higher in the cranial tumor than the caudal. In addition, variation of tumor stroma in a location specific manner also facilitated tumor growth. Cranial sarcoma microenvironment was well vascularized than the caudal one and consequently, a significantly higher microvessel density count was observed which was parallel with low hypoxic response with sign of local tumor inflammation in this region. Taken together, our findings suggest that differential gradient of mitotic regulators together with varied angiogenic response and local tumor microenvironment largely controls solid sarcoma growth along the cranio-caudal anatomical axis.


Aurora Kinase A/biosynthesis , Histones/biosynthesis , Proto-Oncogene Proteins c-myc/biosynthesis , Sarcoma/pathology , Tumor Microenvironment , Animals , Aurora Kinase A/metabolism , Female , Histones/metabolism , Male , Mice , Proto-Oncogene Proteins c-myc/metabolism , Sarcoma/metabolism
...