Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Liver Int ; 42(2): 468-478, 2022 02.
Article in English | MEDLINE | ID: mdl-34719108

ABSTRACT

BACKGROUND AND AIMS: Liver regeneration is a complex process regulated by a variety of cells, cytokines and biological pathways. Aurora kinase A (AURKA) is a serine/threonine kinase that plays a role in centrosome maturation and spindle formation during the cell division cycle. The purpose of this study was to further explore the mechanism of AURKA on liver regeneration and to identify new possible targets for liver regeneration. METHODS: The effect and mechanism of AURKA on liver regeneration were studied using a 70% hepatectomy model. Human liver organoids were used as an in vitro model to investigate the effect of AURKA on hepatocyte proliferation. RESULTS: AURKA inhibition significantly reduced the level of ß-catenin protein by reducing the phosphorylation level of glycogen synthase kinase-3ß (GSK-3ß), leading to the inhibition of liver regeneration. Further studies showed that AURKA co-localized and interacted with GSK-3ß in the cytoplasm of hepatocytes. When phosphorylation of GSK-3ß was enhanced, the total GSK-3ß level remained unchanged, while AURKA was not affected, and ß-catenin protein levels were increased. In addition, AURKA inhibition affected the formation and proliferation of human liver organoids. Furthermore, AURKA inhibition led to the polarization of M1 macrophages and the release of interleukin-6 and Tumour necrosis factor α, which also led to reduced liver regeneration and increased liver injury. CONCLUSIONS: These results provide more details on the mechanism of liver regeneration and suggest that AURKA is an important regulator of this mechanism.


Subject(s)
Aurora Kinase A/physiology , Liver Regeneration , Macrophages/cytology , Wnt Signaling Pathway , Cell Polarity , Glycogen Synthase Kinase 3 beta/physiology , Humans , Macrophages/metabolism , beta Catenin/metabolism
2.
Cell Death Dis ; 12(6): 620, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131100

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid malignancy in children and its mortality rate is relatively high. However, driver genes of NB are not clearly identified. Using bioinformatics analysis, we determined the top 8 differentially expressed genes (DEGs) in NB, including GFAP, PAX6, FOXG1, GAD1, PTPRC, ISL1, GRM5, and GATA3. Insulin gene enhancer binding protein 1 (ISL1) is a LIM homeodomain transcription factor which has been found to be highly expressed in a variety of malignant tumors, but the function of ISL1 in NB has not been fully elucidated. We identified ISL1 as an oncogene in NB. ISL1 is preferentially upregulated in NB tissues compared with normal tissues. High ISL1 expression is significantly associated with poor outcome of NB patients. Knockdown of ISL1 markedly represses proliferation and induces cell apoptosis in vitro, and suppresses tumorigenicity in vivo, while overexpression of ISL1 has the opposite effects. Mechanistically, we demonstrate that ISL1 promotes cell proliferation and EMT transformation through PI3K/AKT signaling pathway by upregulating Aurora kinase A (AURKA), a serine-threonine kinase that is essential for the survival of NB cells. The blockade of AURKA attenuates the function of ISL1 overexpression in the regulation of cell proliferation and migration, Conclusively, this study showed that ISL1 targeted AURKA to facilitate the development of NB, which provided new insights into the tumorigenesis of NB. Thus, ISL1 may be a promising therapeutic target in the future.


Subject(s)
Carcinogenesis/genetics , Epithelial-Mesenchymal Transition/genetics , LIM-Homeodomain Proteins/physiology , Neuroblastoma/genetics , Transcription Factors/physiology , Animals , Aurora Kinase A/metabolism , Aurora Kinase A/physiology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , LIM-Homeodomain Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics
3.
Life Sci Alliance ; 4(6)2021 06.
Article in English | MEDLINE | ID: mdl-33820826

ABSTRACT

Epithelial and haematologic tumours often show the overexpression of the serine/threonine kinase AURKA. Recently, AURKA was shown to localise at mitochondria, where it regulates mitochondrial dynamics and ATP production. Here we define the molecular mechanisms of AURKA in regulating mitochondrial turnover by mitophagy. AURKA triggers the degradation of Inner Mitochondrial Membrane/matrix proteins by interacting with core components of the autophagy pathway. On the inner mitochondrial membrane, the kinase forms a tripartite complex with MAP1LC3 and the mitophagy receptor PHB2, which triggers mitophagy in a PARK2/Parkin-independent manner. The formation of the tripartite complex is induced by the phosphorylation of PHB2 on Ser39, which is required for MAP1LC3 to interact with PHB2. Last, treatment with the PHB2 ligand xanthohumol blocks AURKA-induced mitophagy by destabilising the tripartite complex and restores normal ATP production levels. Altogether, these data provide evidence for a role of AURKA in promoting mitophagy through the interaction with PHB2 and MAP1LC3. This work paves the way to the use of function-specific pharmacological inhibitors to counteract the effects of the overexpression of AURKA in cancer.


Subject(s)
Aurora Kinase A/metabolism , Mitochondria/metabolism , Mitophagy/genetics , Animals , Aurora Kinase A/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster , HEK293 Cells , Humans , MCF-7 Cells , Microtubule-Associated Proteins/metabolism , Mitochondria/physiology , Mitochondrial Dynamics/physiology , Mitochondrial Membranes/metabolism , Mitophagy/physiology , Prohibitins , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases
4.
Nucleic Acids Res ; 48(14): 7844-7855, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32652013

ABSTRACT

The catalytic activity of human AURORA-A kinase (AURKA) regulates mitotic progression, and its frequent overexpression in major forms of epithelial cancer is associated with aneuploidy and carcinogenesis. Here, we report an unexpected, kinase-independent function for AURKA in DNA replication initiation whose inhibition through a class of allosteric inhibitors opens avenues for cancer therapy. We show that genetic depletion of AURKA, or its inhibition by allosteric but not catalytic inhibitors, blocks the G1-S cell cycle transition. A catalytically inactive AURKA mutant suffices to overcome this block. We identify a multiprotein complex between AURKA and the replisome components MCM7, WDHD1 and POLD1 formed during G1, and demonstrate that allosteric but not catalytic inhibitors prevent the chromatin assembly of functional replisomes. Indeed, allosteric but not catalytic AURKA inhibitors sensitize cancer cells to inhibition of the CDC7 kinase subunit of the replication-initiating factor DDK. Thus, our findings define a mechanism essential for replisome assembly during DNA replication initiation that is vulnerable to inhibition as combination therapy in cancer.


Subject(s)
Aurora Kinase A/physiology , DNA Replication , DNA-Directed DNA Polymerase/metabolism , Multienzyme Complexes/metabolism , Allosteric Regulation , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Line , G1 Phase Cell Cycle Checkpoints , HeLa Cells , Humans , Interphase/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Replication Origin
5.
Int J Oncol ; 57(5): 1095-1102, 2020 11.
Article in English | MEDLINE | ID: mdl-33491761

ABSTRACT

The primary cilium is a non­motile cellular structure extending from the apical membrane of epithelial cells that is involved in several processes due to its ability to receive and elaborate different signals. Ciliogenesis and its obliteration are essential for proliferating cells, and several signalling pathways are responsible for their regulation. In fact, the primary cilium is a central hub for numerous signalling pathways implicated in a variety of biological processes, such as the Hedgehog, mammalian target of rapamycin and Wnt pathways. Loss of primary cilia has been recently correlated with different types of tumours, including pancreatic ductal adenocarcinoma (PDAC). K­Ras and HDAC2 were recently identified as possible ciliogenesis regulators in PDAC, likely acting through Aurora A kinase (AURKA) which, in turn, controls inositol polyphosphate­5­phosphatase E. However, the exact molecular mechanisms underlying this regulatory effect remain to be fully elucidated. In the present study, the regulation of the main genes involved in primary cilia assembly/resorption was reconstructed showing the links with the Hedgehog and phosphoinositide 3­kinase/AKT pathways. Finally, by analysing gene expression databases, the regulatory genes that have high probability to be associated with prognosis, histological grade and pathological stage in patients with PDAC have been highlighted. However, further experimental studies are required to reach definitive conclusions on the roles of these genes. Improving our understating of ciliogenesis and its regulators may help develop ciliotherapies using histone deacetylase and AURKA inhibitors, which may induce re­differentiation of tumour cells into normal cells by reducing tumour growth or inducing apoptosis of cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cilia/physiology , Pancreatic Neoplasms/pathology , Aurora Kinase A/physiology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cilia/drug effects , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/therapeutic use , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction
6.
Oncogene ; 38(1): 73-87, 2019 01.
Article in English | MEDLINE | ID: mdl-30082913

ABSTRACT

Primary glioblastoma is the most frequent human brain tumor in adults and is generally fatal due to tumor recurrence. We previously demonstrated that glioblastoma-initiating cells invade the subventricular zones and promote their radio-resistance in response to the local release of the CXCL12 chemokine. In this work, we show that the mitotic Aurora A kinase (AurA) is activated through the CXCL12-CXCR4 pathway in an ERK1/2-dependent manner. Moreover, the CXCL12-ERK1/2 signaling induces the expression of Ajuba, the main cofactor of AurA, which allows the auto-phosphorylation of AurA.We show that AurA contributes to glioblastoma cell survival, radio-resistance, self-renewal, and proliferation regardless of the exogenous stimulation with CXCL12. On the other hand, AurA triggers the CXCL12-mediated migration of glioblastoma cells in vitro as well as the invasion of the subventricular zone in xenograft experiments. Moreover, AurA regulates cytoskeletal proteins (i.e., Actin and Vimentin) and favors the pro-migratory activity of the Rho-GTPase CDC42 in response to CXCL12. Altogether, these results show that AurA, a well-known kinase of the mitotic machinery, may play alternative roles in human glioblastoma according to the CXCL12 concentration.


Subject(s)
Aurora Kinase A/physiology , Brain Neoplasms/enzymology , Chemokine CXCL12/physiology , Glioblastoma/enzymology , Neoplasm Proteins/physiology , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival , Chemokine CXCL12/pharmacology , Enzyme Activation , Glioblastoma/pathology , Heterografts , Humans , LIM Domain Proteins/biosynthesis , LIM Domain Proteins/genetics , Lateral Ventricles/pathology , MAP Kinase Signaling System , Mice , Neoplasm Invasiveness , Phosphorylation , Protein Processing, Post-Translational , Receptors, CXCR4/physiology , Signal Transduction
7.
Dev Biol ; 440(2): 88-98, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29753017

ABSTRACT

In metazoans, organisms arising from a fertilized egg, the embryo will develop through multiple series of cell divisions, both symmetric and asymmetric, leading to differentiation. Aurora A is a serine threonine kinase highly involved in such divisions. While intensively studied at the cell biology level, its function in the development of a whole organism has been neglected. Here we investigated the pleiotropic effect of Aurora A loss-of-function in Drosophila larval early development. We report that Aurora A is required for proper larval development timing control through direct and indirect means. In larval tissues, Aurora A is required for proper symmetric division rate and eventually development speed as we observed in central brain, wing disc and ring gland. Moreover, Aurora A inactivation induces a reduction of ecdysteroids levels and a pupariation delay as an indirect consequence of ring gland development deceleration. Finally, although central brain development is initially restricted, we confirmed that brain lobe size eventually increases due to additive phenotypes: delayed pupariation and over-proliferation of cells with an intermediate cell-identity between neuroblast and ganglion mother cell resulting from defective asymmetric neuroblast cell division.


Subject(s)
Aurora Kinase A/physiology , Drosophila Proteins/physiology , Drosophila/embryology , Larva/metabolism , Animals , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Brain/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Division/physiology , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Genetic Pleiotropy/genetics , Larva/physiology , Loss of Function Mutation/genetics , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neurons/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/physiology , Spindle Apparatus/metabolism
8.
Mol Cells ; 41(5): 444-453, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29477140

ABSTRACT

Aberrations in histone modifications are being studied in mixed-lineage leukemia (MLL)-AF9-driven acute myeloid leukemia (AML). In this study, we focused on the regulation of the differentiation of the MLL-AF9 type AML cell line THP-1. We observed that, upon phorbol 12-myristate 13-acetate (PMA) treatment, THP-1 cells differentiated into monocytes by down-regulating Aurora kinase A (AURKA), resulting in a reduction in H3S10 phosphorylation. We revealed that the AURKA inhibitor alisertib accelerates the expression of the H3K27 demethylase KDM6B, thereby dissociating AURKA and YY1 from the KDM6B promoter region. Using Flow cytometry, we found that alisertib induces THP-1 differentiation into monocytes. Furthermore, we found that treatment with the KDM6B inhibitor GSK-J4 perturbed the PMA-mediated differentiation of THP-1 cells. Thus, we discovered the mechanism of AURKA-KDM6B signaling that controls the differentiation of THP-1 cells, which has implications for biotherapy for leukemia.


Subject(s)
Aurora Kinase A/physiology , Gene Expression Regulation, Leukemic , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/physiology , Leukemia, Monocytic, Acute/pathology , Neoplasm Proteins/physiology , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Benzazepines/pharmacology , Cell Differentiation/drug effects , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Genes, Reporter , HEK293 Cells , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Leukemia, Monocytic, Acute/genetics , Leukemia, Monocytic, Acute/metabolism , Monocytes/cytology , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/antagonists & inhibitors , Oncogene Proteins, Fusion/physiology , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Processing, Post-Translational/drug effects , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology , YY1 Transcription Factor/metabolism
9.
Nat Rev Cancer ; 17(2): 93-115, 2017 01 27.
Article in English | MEDLINE | ID: mdl-28127048

ABSTRACT

Cancer is characterized by uncontrolled tumour cell proliferation resulting from aberrant activity of various cell cycle proteins. Therefore, cell cycle regulators are considered attractive targets in cancer therapy. Intriguingly, animal models demonstrate that some of these proteins are not essential for proliferation of non-transformed cells and development of most tissues. By contrast, many cancers are uniquely dependent on these proteins and hence are selectively sensitive to their inhibition. After decades of research on the physiological functions of cell cycle proteins and their relevance for cancer, this knowledge recently translated into the first approved cancer therapeutic targeting of a direct regulator of the cell cycle. In this Review, we focus on proteins that directly regulate cell cycle progression (such as cyclin-dependent kinases (CDKs)), as well as checkpoint kinases, Aurora kinases and Polo-like kinases (PLKs). We discuss the role of cell cycle proteins in cancer, the rationale for targeting them in cancer treatment and results of clinical trials, as well as the future therapeutic potential of various cell cycle inhibitors.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/physiology , Cell Cycle Proteins/physiology , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/physiology , Clinical Trials as Topic , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/physiology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/physiology , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , Signal Transduction , Polo-Like Kinase 1
10.
Nat Commun ; 7: 11727, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27242098

ABSTRACT

The Golgi apparatus is composed of stacks of cisternae laterally connected by tubules to form a ribbon-like structure. At the onset of mitosis, the Golgi ribbon is broken down into discrete stacks, which then undergo further fragmentation. This ribbon cleavage is required for G2/M transition, which thus indicates that a 'Golgi mitotic checkpoint' couples Golgi inheritance with cell cycle transition. We previously showed that the Golgi-checkpoint regulates the centrosomal recruitment of the mitotic kinase Aurora-A; however, how the Golgi unlinking regulates this recruitment was unknown. Here we show that, in G2, Aurora-A recruitment is promoted by activated Src at the Golgi. Our data provide evidence that Src and Aurora-A interact upon Golgi ribbon fragmentation; Src phosphorylates Aurora-A at tyrosine 148 and this specific phosphorylation is required for Aurora-A localization at the centrosomes. This process, pivotal for centrosome maturation, is a fundamental prerequisite for proper spindle formation and chromosome segregation.


Subject(s)
Aurora Kinase A/physiology , Centrosome/physiology , G2 Phase/physiology , Golgi Apparatus/metabolism , src-Family Kinases/physiology , Animals , Aurora Kinase A/genetics , CSK Tyrosine-Protein Kinase , Chromosome Segregation/physiology , HeLa Cells , Humans , Indoles/pharmacology , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Rats , S Phase/drug effects , Sulfonamides/pharmacology , Thymidine/pharmacology , Tyrosine/metabolism , src-Family Kinases/antagonists & inhibitors
11.
Sci Rep ; 6: 28436, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27341528

ABSTRACT

AurkA overexpression was previously found in breast cancer and associated to its ability in controlling chromosome segregation during mitosis, however whether it may affect breast cancer cells, endorsed with stem properties (BCICs), is still unclear. Surprisingly, a strong correlation between AurkA expression and ß-catenin localization in breast cancer tissues suggested a link between AurkA and Wnt signaling. In our study, AurkA knock-down reduced wnt3a mRNA and suppressed metastatic signature of MDA-MB-231 cells. As a consequence, the amount of BCICs and their migratory capability dramatically decreased. Conversely, wnt3a mRNA stabilization and increased CD44(+)/CD24(low/-) subpopulation was found in AurkA-overexpressing MCF7 cells. In vivo, AurkA-overexpressing primary breast cancer cells showed higher tumorigenic properties. Interestingly, we found that AurkA suppressed the expression of miR-128, inhibitor of wnt3a mRNA stabilization. Namely, miR-128 suppression realized after AurkA binding to Snail. Remarkably, a strong correlation between AurkA and miR-128 expression in breast cancer tissues confirmed our findings. This study provides novel insights into an undisclosed role for the kinase AurkA in self-renewal and migration of BCICs affecting response to cancer therapies, metastatic spread and recurrence. In addition, it suggests a new therapeutic strategy taking advantage of miR-128 to suppress AurkA-Wnt3a signaling.


Subject(s)
Aurora Kinase A/physiology , Breast Neoplasms/enzymology , MicroRNAs/genetics , Neoplastic Stem Cells/physiology , Wnt3A Protein/metabolism , Active Transport, Cell Nucleus , Animals , Breast Neoplasms/pathology , Cell Self Renewal , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Mice , MicroRNAs/metabolism , Neoplasm Transplantation , Protein Stability , beta Catenin/metabolism
12.
Clin Lab ; 62(4): 697-703, 2016.
Article in English | MEDLINE | ID: mdl-27215090

ABSTRACT

BACKGROUND: Suppression of Aurora kinase A (Aurora-A, AURKA) by Aurora-A siRNA has been proposed for lung tumor treatment. However, protocols using single administration have shown little benefit in some types of lung tumor. Given that transfection efficiency of Aurora-A siRNA is low due to tightly packed cells in the tumor, we hypothesized that repeated administration would result in efficient cell apoptosis. METHODS: We compared single vs. repeated transfection (thrice) in A549 cells by transfecting Aurora-A siRNA (siA) on the 1st or 1st, 2nd and 3rd day after cell seeding. A random sequence was used as the negative siRNA control (siC). Cells in the single transfection group received only transfection reagent without siRNAs on the 2nd and 3rd day. RESULTS: Two days after the third transfection, both single and repeated siA administration decreased mRNA expression of Aurora-A and cell viability compared to no administration and siC single administration. However, the decrease in these two indices with repeated transfection was more obvious than that following single administration: cell viability decreased to 72.8 ± 3.05% (p < 0.05) following siA single transfection and to 64.2 ± 1.99% (p < 0.05) following siA repeated transfection, compared with normal control cells, respectively. Gene expression decreased to 17 ± 16.6% (p < 0.05 vs. normal control) following siA repeated transfection and to 43.2 ± 13.0% (p < 0.05 vs. normal control) following siA single transfection. CONCLUSIONS: Compared to single transfection, repeated Aurora-A siRNA transfection decreased Aurora-A, which, in turn, resulted in effective apoptosis of A549 cells.


Subject(s)
Apoptosis , Aurora Kinase A/genetics , RNA, Small Interfering/genetics , Transfection , Aurora Kinase A/physiology , Cell Line, Tumor , Cell Survival , Humans
13.
Cancer Cell ; 29(4): 536-547, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27050099

ABSTRACT

MYCN amplification and overexpression are common in neuroendocrine prostate cancer (NEPC). However, the impact of aberrant N-Myc expression in prostate tumorigenesis and the cellular origin of NEPC have not been established. We define N-Myc and activated AKT1 as oncogenic components sufficient to transform human prostate epithelial cells to prostate adenocarcinoma and NEPC with phenotypic and molecular features of aggressive, late-stage human disease. We directly show that prostate adenocarcinoma and NEPC can arise from a common epithelial clone. Further, N-Myc is required for tumor maintenance, and destabilization of N-Myc through Aurora A kinase inhibition reduces tumor burden. Our findings establish N-Myc as a driver of NEPC and a target for therapeutic intervention.


Subject(s)
Adenocarcinoma/genetics , Cell Transformation, Neoplastic/genetics , Epithelial Cells/pathology , Neoplasm Proteins/physiology , Neuroendocrine Tumors/genetics , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/physiology , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/therapeutic use , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/physiology , Azepines/therapeutic use , Cell Line, Tumor , Enzyme Activation , Epithelial Cells/metabolism , Exome , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Laser Capture Microdissection , Male , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neuroendocrine Tumors/pathology , Orchiectomy , Phenylurea Compounds/therapeutic use , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/physiology , Pyrimidines/therapeutic use , Recombinant Fusion Proteins/metabolism , Transduction, Genetic , Xenograft Model Antitumor Assays
14.
Tumour Biol ; 37(3): 3071-80, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26423403

ABSTRACT

Colorectal adenomatous polyp (CRAP) is a major risk factor for the development of sporadic colorectal cancer (CRC). Histone modifications are one of the epigenetic mechanisms that may have key roles in the carcinogenesis of CRC. The objective of the present study is to investigate the alternations in the defined histone modification gene expression profiles in patients with CRAP and CRC. Histone modification enzyme key gene expressions of the CRC, CRAP, and control groups were evaluated and compared using the reverse transcription PCR (RT-PCR) array method. Gene expression analysis was performed in the CRAP group after dividing the patients into subgroups according to the polyp diameter, pathological results, and morphological parameters which are risk factors for developing CRC in patients with CRAP. PAK1, NEK6, AURKA, AURKB, HDAC1, and HDAC7 were significantly more overexpressed in CRC subjects compared to the controls (p < 0.05). PAK1, NEK6, AURKA, AURKB, and HDAC1 were significantly more overexpressed in the CRAP group compared to the controls (p < 0.005). There were no significant differences between the CRAP and CRC groups with regards to PAK1, NEK6, AURKA, or AURKB gene overexpression. PAK1, NEK6, AURKA, and AURKB were significantly in correlation with the polyp diameter as they were more overexpressed in polyps with larger diameters. In conclusion, overexpressions of NEK6, AURKA, AURKB, and PAK1 genes can be used as predictive markers to decide the colonoscopic surveillance intervals after the polypectomy procedure especially in polyps with larger diameters.


Subject(s)
Adenocarcinoma/genetics , Adenomatous Polyposis Coli/genetics , Aurora Kinase A/genetics , Aurora Kinase B/genetics , Colorectal Neoplasms/genetics , p21-Activated Kinases/genetics , Adenocarcinoma/pathology , Adenomatous Polyposis Coli/pathology , Adult , Aged , Aurora Kinase A/physiology , Aurora Kinase B/physiology , Colorectal Neoplasms/pathology , Female , Humans , Male , Middle Aged , NIMA-Related Kinases/genetics , NIMA-Related Kinases/physiology , p21-Activated Kinases/physiology
15.
J Cell Biol ; 210(1): 45-62, 2015 Jul 06.
Article in English | MEDLINE | ID: mdl-26124292

ABSTRACT

Coordination of cell growth and proliferation in response to nutrient supply is mediated by mammalian target of rapamycin (mTOR) signaling. In this study, we report that Mio, a highly conserved member of the SEACAT/GATOR2 complex necessary for the activation of mTORC1 kinase, plays a critical role in mitotic spindle formation and subsequent chromosome segregation by regulating the proper concentration of active key mitotic kinases Plk1 and Aurora A at centrosomes and spindle poles. Mio-depleted cells showed reduced activation of Plk1 and Aurora A kinase at spindle poles and an impaired localization of MCAK and HURP, two key regulators of mitotic spindle formation and known substrates of Aurora A kinase, resulting in spindle assembly and cytokinesis defects. Our results indicate that a major function of Mio in mitosis is to regulate the activation/deactivation of Plk1 and Aurora A, possibly by linking them to mTOR signaling in a pathway to promote faithful mitotic progression.


Subject(s)
Aurora Kinase A/physiology , Cell Cycle Proteins/metabolism , Centrosome/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Enzyme Activation , Gene Knockdown Techniques , HeLa Cells , Humans , Kinesins/metabolism , Mitosis , Molecular Sequence Data , Neoplasm Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Structure, Tertiary , Protein Transport , Spindle Apparatus/metabolism , Polo-Like Kinase 1
16.
Blood ; 125(13): 2141-50, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25670627

ABSTRACT

Aurora kinase A (AURKA) is a therapeutic target in acute megakaryocytic leukemia. However, its requirement in normal hematopoiesis and megakaryocyte development has not been extensively characterized. Based on its role as a cell cycle regulator, we predicted that an Aurka deficiency would lead to severe abnormalities in all hematopoietic lineages. Here we reveal that loss of Aurka in hematopoietic cells causes profound cell autonomous defects in the peripheral blood and bone marrow. Surprisingly, in contrast to the survival defects of nearly all hematopoietic lineages, deletion of Aurka was associated with increased differentiation and polyploidization of megakaryocytes both in vivo and in vitro. Furthermore, in contrast to other cell types examined, megakaryocytes continued DNA synthesis after loss of Aurka. Thus, like other cell cycle regulators such as Aurkb and survivin, Aurka is required for hematopoiesis, but is dispensable for megakaryocyte endomitosis. Our work supports a growing body of evidence that the megakaryocyte endomitotic cell cycle differs significantly from the proliferative cell cycle.


Subject(s)
Aurora Kinase A/physiology , Cell Differentiation/genetics , Hematopoiesis/genetics , Megakaryocytes/physiology , Mitosis/genetics , Adult Stem Cells/physiology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , Polyploidy , Thrombopoiesis/genetics
17.
Oncol Rep ; 33(4): 1860-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25634113

ABSTRACT

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (EGFR-TKIs) have been used to treat non-small cell lung carcinoma (NSCLC) patients that have EGFR-activating mutations. EGFR-TKI monotherapy in most NSCLC patients with EGFR mutations who initially respond to EGFR-TKIs results in the development of acquired resistance. We investigated the role of fibroblasts in stromal cell-mediated resistance to gefitinib-induced apoptosis in EGFR-mutant NSCLC cells. While gefitinib induced apoptosis in EGFR-mutant NSCLC cells, apoptosis induction was diminished under stromal co-culture conditions. Protection appeared to be mediated in part by Aurora-A kinase (AURKA) upregulation. The protective effect of stromal cells was significantly reduced by pre-exposure to AURKA-shRNA. We suggest that combinations of AURKA antagonists and EGFR inhibitors may be effective in clinical trials targeting mutant EGFR NSCLCs.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinase A/physiology , Carcinoma, Non-Small-Cell Lung/pathology , Fibroblasts/enzymology , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/pathology , Neoplasm Proteins/physiology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Aurora Kinase A/biosynthesis , Aurora Kinase A/genetics , Carcinoma, Non-Small-Cell Lung/enzymology , Cell Line, Tumor , Coculture Techniques , Drug Resistance, Neoplasm/physiology , Enzyme Induction/drug effects , ErbB Receptors/deficiency , Gefitinib , Gene Regulatory Networks/drug effects , Genes, erbB-1 , Humans , Lung Neoplasms/enzymology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Signal Transduction , Stromal Cells/enzymology , Tumor Suppressor Protein p53/physiology , Up-Regulation/drug effects
18.
Inflammation ; 38(2): 800-11, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25227280

ABSTRACT

Macrophage polarization is a dynamic and integral process of tissue inflammation and remodeling. Here we demonstrate an important role of Aurora kinase A in the regulation of inflammatory M1 macrophage polarization. We found that there was an elevated expression of Aurora-A in M1 macrophages and inhibition of Aurora-A by small molecules or specific siRNA selectively led to the suppression of M1 polarization, sparing over the M2 macrophage differentiation. At the molecular level, we found that the effects of Aurora-A in M1 macrophages were mediated through the down-regulation of NF-κB pathway and subsequent IRF5 expression. In an autoimmune disease model, experimental autoimmune encephalitis (EAE), treatment with Aurora kinase inhibitor blocked the disease development and shifted the macrophage phenotype from inflammatory M1 to anti-inflammatory M2. Thus, this study reveals a novel function of Aurora-A in controlling the polarization of macrophages, and modification of Aurora-A activity may lead to a new therapeutic approach for chronic inflammatory diseases.


Subject(s)
Aurora Kinase A/physiology , Cell Polarity/physiology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Macrophages/enzymology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Polarity/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Humans , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use
19.
J BUON ; 20(6): 1414-9, 2015.
Article in English | MEDLINE | ID: mdl-26854435

ABSTRACT

PURPOSE: The aim of this study was to determine the expression level of Aurora A in human breast cancer tissues and to test whether there is a relationship between its expression levels and clinicopathological parameters including response to taxanes, tumor grade, estrogen receptor (ER) status, human epidermal growth factor receptor 2 (HER2) status, and overall survival (OS). METHODS: We retrospectively analyzed paraffin-embedded tissue sections from 49 metastatic breast cancer patients whose clinical outcomes had been tracked after taxane treatment. The expression status of Aurora A was defined by immunohistochemistry (IHC) using the anti-Aurora A antibody. RESULTS: Aurora A was overexpressed in 73% of the examined specimens. There was significant correlation between high Aurora A expression and decreased taxane sensitivity (p=0.02). There was no association between the clinicopathological parameters including histologic grade, ER positivity and triple negative molecular subtype and the level of Aurora A expression. However, HER2 positive tumors showed significantly higher Aurora A expression compared with HER2 negative tumors (p=0.02). Kaplan-Meier survival analysis failed to show a significant correlation between expression levels of Aurora A and OS although patients with low Aurora A levels had a marginally longer survival compared to patients with high levels. CONCLUSION: Our data suggest that Aurora A may be a promising predictive and prognostic marker in patients with breast cancer.


Subject(s)
Aurora Kinase A/physiology , Breast Neoplasms/enzymology , Bridged-Ring Compounds/therapeutic use , Taxoids/therapeutic use , Aurora Kinase A/analysis , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Drug Resistance, Neoplasm , Female , Humans , Prognosis , Receptor, ErbB-2/analysis , Retrospective Studies
20.
Biochim Biophys Acta ; 1846(2): 630-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25450825

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is an aggressive variant of prostate cancer that commonly arises in later stages of castration resistant prostate cancer (CRPC) The detection of NEPC has clinical implications as these patients are often treated with platinum chemotherapy rather than with androgen receptor targeted therapies. The poor molecular characterization of NEPC accounts in part for the lack of disease specific therapeutics. Several mechanisms are involved in NE differentiation, including inflammation and autophagy, and may actually represent future therapeutic targets for advanced NEPC patients. Furthermore, a growing body of evidence suggests a potential role of circulating tumor cells in the early diagnosis and treatment of NEPC. Here we summarize the recent findings on NEPC pathogenesis and we discuss the ongoing clinical trials and future perspectives for the treatment of NEPC patients.


Subject(s)
Neuroendocrine Cells/cytology , Prostatic Neoplasms/pathology , Angiogenesis Inhibitors/therapeutic use , Aurora Kinase A/physiology , Autophagy , Carcinogenesis , Cell Differentiation , Drug Resistance, Neoplasm , Humans , Male , Neoplastic Cells, Circulating , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...