Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Cell Biochem ; 122(7): 739-751, 2021 07.
Article in English | MEDLINE | ID: mdl-33586155

ABSTRACT

The appropriate regulation of spindle orientation maintains proper tissue homeostasis and avoids aberrant tissue repair or regeneration. Spindle misorientation due to imbalance or improper functioning leads to a loss of tissue integrity and aberrant growth, such as tissue loss or overgrowth. Pharmacological manipulation to prevent spindle misorientation will enable a better understanding of how spindle orientation is involved in physiological and pathological conditions and will provide therapeutic possibilities to treat patients associated with abnormal tissue function caused by spindle misorientation. N-terminal-deleted Rho guanine nucleotide dissociation inhibitor Ɵ (RhoGDIƟ/RhoGDI2/LyGDI) produced by caspase-3 activation perturbs spindle orientation in surviving cells following exposure to either ionizing radiation or UVC. Thus, presumably, RhoGDIƟ cleaved by caspase-3 activation acts as a determinant of radiation-induced spindle misorientation that promote aberrant tissue repair due to deregulation of directional organization of cell population and therefore becomes a potential target of drugs to prevent such response. The objective of this study was to screen and identify chemicals that suppress RhoGDIƟ expression. We focused our attention on ascorbic acid (AA) derivatives because of their impact on the maintenance of skin tissue homeostasis. Here, we screened for AA derivatives that suppress RhoGDIƟ expression in HeLa cells and identified a lipophilic derivative, 2-O-octadecylascorbic acid (2-OctadecylAA), as a novel RhoGDIƟ inhibitor that ameliorated ionizing radiation-induced abnormal spindle orientations. Among all examined AA derivatives, which were also antioxidative, the inhibition activity was specific to 2-OctadecylAA. Therefore, this activity was not due to simple antioxidant properties. 2-OctadecylAA was previously shown to prevent hepatocellular carcinoma development. Our findings suggest that the anticarcinogenic effects of 2-OctadecylAA are partly due to RhoGDIƟ inhibition mechanisms by which spindle orientation perturbations are attenuated. Thus, the molecular targeting features of RhoGDIƟ warrant its further development for the treatment or control of spindle orientation abnormalities that affect epithelial homeostasis.


Subject(s)
Ascorbic Acid/analogs & derivatives , DNA Damage , Gene Expression Regulation/drug effects , Spindle Apparatus/pathology , rho Guanine Nucleotide Dissociation Inhibitor beta/antagonists & inhibitors , Ascorbic Acid/pharmacology , HeLa Cells , Humans , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism
2.
J Cell Physiol ; 234(9): 15134-15146, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30652309

ABSTRACT

The molecular signals that regulate mitotic spindle orientation to determine the proper division axis play a critical role in the development and maintenance of tissue homeostasis. However, deregulation of signaling events can result in spindle misorientation, which in turn can trigger developmental defects and cancer progression. Little is known about the cellular signaling pathway involved in the misorientation of proliferating cells that evade apoptosis after DNA damage. In this study, we found that perturbations to spindle orientation were induced in ultraviolet C (UVC)-irradiated surviving cells. N-terminal truncated Rho GDP-dissociation inhibitor Ɵ (RhoGDIƟ), which is produced by UVC irradiation, distorted the spindle orientation of HeLa cells cultured on Matrigel. The short hairpin RNA-mediated knockdown of RhoGDIƟ significantly attenuated UVC-induced misorientation. Subsequent expression of wild-type RhoGDIƟ, but not a noncleavable mutant, RhoGDIƟ (D19A), again led to a relative increase in spindle misorientation in response to UVC. Our findings revealed that RhoGDIƟ impacts spindle orientation in response to DNA damage.

3.
PLoS Genet ; 10(9): e1004639, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25233213

ABSTRACT

Nonessential tRNA modifications by methyltransferases are evolutionarily conserved and have been reported to stabilize mature tRNA molecules and prevent rapid tRNA decay (RTD). The tRNA modifying enzymes, NSUN2 and METTL1, are mammalian orthologs of yeast Trm4 and Trm8, which are required for protecting tRNA against RTD. A simultaneous overexpression of NSUN2 and METTL1 is widely observed among human cancers suggesting that targeting of both proteins provides a novel powerful strategy for cancer chemotherapy. Here, we show that combined knockdown of NSUN2 and METTL1 in HeLa cells drastically potentiate sensitivity of cells to 5-fluorouracil (5-FU) whereas heat stress of cells revealed no effects. Since NSUN2 and METTL1 are phosphorylated by Aurora-B and Akt, respectively, and their tRNA modifying activities are suppressed by phosphorylation, overexpression of constitutively dephosphorylated forms of both methyltransferases is able to suppress 5-FU sensitivity. Thus, NSUN2 and METTL1 are implicated in 5-FU sensitivity in HeLa cells. Interfering with methylation of tRNAs might provide a promising rationale to improve 5-FU chemotherapy of cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Methyltransferases/metabolism , RNA, Transfer/metabolism , Cell Proliferation , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression , Gene Knockdown Techniques , HeLa Cells , Heat-Shock Response/genetics , Humans , Methyltransferases/genetics , Paclitaxel/pharmacology , Phosphorylation , RNA Stability/drug effects , RNA, Transfer/genetics , Tumor Stem Cell Assay
4.
Carcinogenesis ; 37(10): 993-1003, 2016 10.
Article in English | MEDLINE | ID: mdl-27515963

ABSTRACT

The mitotic Aurora B kinase is overexpressed in tumors and various inhibitors for Aurora B are currently under clinical assessments. However, when considering Aurora B kinase inhibitors as anticancer drugs, their mode of action and the role of p53 status as a possible predictive factor for response still needs to be investigated. In this study, we analyzed the effects of selective Aurora B inhibition using AZD1152-HQPA/Barasertib (AZD1152) on HCT116 cells, U87-MG, corresponding isogenic p53-deficient cells and a primary glioblastoma cell line. AZD1152 treatment caused polyploidy and non-apoptotic cell death in all cell lines irrespective of p53 status and was accompanied by poly-merotelic kinetochore-microtubule attachments and DNA damage. In p53 wild-type cells a DNA damage response induced an inefficient pseudo-G1 cell cycle arrest, which was not able to halt ongoing endoreplication of cells. Of note, release of tumor cells from AZD1152 resulted in recovery of aneuploid progenies bearing numerical and structural chromosomal aberrations. Yet, AZD1152 treatment enhanced death receptor TRAIL-R2 levels in all tumor cell lines investigated. A concomitant increase of the activating natural killer (NK) cell ligand MIC A/B in p53-deficient cells and an induction of FAS/CD95 in cells containing p53 rendered AZD1152-treated cells more susceptible for NK-cell-mediated lysis. Our study mechanistically explains a p53-independent mode of action of a chemical Aurora B inhibitor and suggests a potential triggering of antitumoral immune responses, following polyploidization of tumor cells, which might constrain recovery of aneuploid tumor cells.


Subject(s)
Apoptosis/drug effects , Aurora Kinase B/biosynthesis , Cell Proliferation/drug effects , Tumor Suppressor Protein p53/genetics , Aneuploidy , Aurora Kinase B/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Chromosome Aberrations/drug effects , DNA Damage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Organophosphates/administration & dosage , Quinazolines/administration & dosage , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Suppressor Protein p53/biosynthesis
5.
J Cell Physiol ; 231(11): 2493-505, 2016 11.
Article in English | MEDLINE | ID: mdl-26919575

ABSTRACT

The equilibrium between proliferation and apoptosis is tightly balanced to maintain tissue homeostasis in normal tissues and even in tumors. Achieving and maintaining such a balance is important for cancer regrowth and spreading after cytotoxic treatments. Caspase-3 activation and tumor cell death following anticancer therapy as well as accompanying cell death pathways are well characterized, but their association to homeostasis of cancerous tissue and tumor progression remains poorly understood. Here we proposed a novel mechanism of cancer spreading induced by caspase-3. RhoGDIƟ, known as a direct cleavage substrate of caspase-3, is overexpressed in many epithelial cancers. The N-terminal-truncated RhoGDIƟ (ΔN-RhoGDIƟ) is accumulated in caspase-3-activated cells. Stable expression of ΔN-RhoGDIƟ in HeLa cells did not induce apoptosis, but impaired directional cell migration in a wound-healing assay accompanied by a perturbed direction of cell division at the wound edge. Subcellular protein fractionation experiments revealed that ΔN-RhoGDIƟ but not wild-type RhoGDIƟ was present in the detergent-soluble cytoplasmic and nuclear fractions and preferentially associated with Cdc42. Furthermore, Cdc42 activity was constitutively inhibited by stable expression of ΔN-RhoGDIƟ, resulting in increased radiation-induced compensatory proliferation linking to RhoA activation. Thus, ΔN-RhoGDIƟ dominant-negatively regulates Cdc42 activity and contributes to loss of polarity-related functions. The caspase-3-cleaved RhoGDIƟ is a possible determinant to promote cancer spreading due to deregulation of directional organization of tumor cell population and inhibition of default equilibrium between proliferation and apoptosis after cytotoxic damage. J. Cell. Physiol. 231: 2493-2505, 2016. Ā© 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Movement/radiation effects , Cell Polarity/radiation effects , Neoplasms/pathology , Radiation , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Division/radiation effects , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Down-Regulation/radiation effects , Enzyme Activation/radiation effects , Genes, Dominant , HeLa Cells , Humans , Models, Biological , Mutant Proteins/metabolism , Neoplasm Metastasis , Protein Transport/radiation effects , Subcellular Fractions/metabolism , X-Rays , cdc42 GTP-Binding Protein/metabolism
6.
Mol Cancer ; 13: 107, 2014 May 09.
Article in English | MEDLINE | ID: mdl-24886358

ABSTRACT

BACKGROUND: Survivin, a member of the inhibitor of apoptosis (IAP) gene family, has a dual role in mitosis and in apoptosis. It is abundantly expressed in every human tumor, compared with normal tissues. During mitosis Survivin assembles with the chromosomal passenger complex and regulates chromosomal segregation. Here, we aim to explore whether interference with the mitotic function of Survivin is linked to p53-mediated G1 cell cycle arrest and affects chromosomal stability. METHODS: In this study, we used HCT116, SBC-2, and U87-MG and generated corresponding isogenic p53-deficient cells. Retroviral vectors were used to stably knockdown Survivin. The resulting phenotype, in particular the mechanisms of cell cycle arrest and of initiation of aneuploidy, were investigated by Western Blot analysis, confocal laser scan microscopy, proliferation assays, spectral karyotyping and RNAi. RESULTS: In all cell lines Survivin-RNAi did not induce instant apoptosis but caused polyplodization irrespective of p53 status. Strikingly, polyploidization after knockdown of Survivin resulted in merotelic kinetochore spindle assemblies, ƎĀ³H2AX-foci, and DNA damage response (DDR), which was accompanied by a transient p53-mediated G1-arrest. That p53 wild type cells specifically arrest due to DNA damage was shown by simultaneous inhibition of ATM and DNA-PK, which abolished induction of p21waf/cip. Cytogenetic analysis revealed chromosomal aberrations indicative for DNA double strand break repair by the mechanism of non-homologous end joining (NHEJ), only in Survivin-depleted cells. CONCLUSION: Our findings suggest that Survivin plays an essential role in proper amphitelic kinetochore-spindle assembly and that constraining Survivin's mitotic function results in polyploidy and aneuploidy which cannot be controlled by p53. Therefore, Survivin critically safeguards chromosomal stability independently from p53.


Subject(s)
Aneuploidy , DNA End-Joining Repair , Inhibitor of Apoptosis Proteins/genetics , Mitosis , Polyploidy , Tumor Suppressor Protein p53/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Chromosome Segregation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Breaks, Double-Stranded , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Gene Expression Regulation , Genomic Instability , Histones/genetics , Histones/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Karyotyping , Kinetochores/metabolism , Kinetochores/ultrastructure , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Survivin , Tumor Suppressor Protein p53/metabolism
7.
Mutat Res ; 688(1-2): 78-87, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20346366

ABSTRACT

In mouse thymic lymphoma 3SB cells bearing wild type p53, ionizing radiation (IR) and UV light are potent triggers of caspase-3-dependent apoptosis. Although cytochrome c was released from mitochondria as expected, caspase-9 activation was not observed in UV-exposed cells. Laser scanning confocal microscopy analysis showed that caspase-9 is localized in an unusual punctuated pattern in UV-induced apoptotic cells. In agreement with differences in the status of caspase-9 activation between IR and UV, subcellular protein fractionation experiments showed that pro-apoptotic apoptosis protease-activating factor 1 (Apaf-1), normally a part of the apoptosome assembled in response to the release of cytochrome c from mitochondria, and B-cell lymphoma extra long (Bcl-xL), an inhibitor of the change in mitochondrial membrane permeability, were redistributed by the IR-exposure but not by the UV-exposure. Instead of the sequestration of the capase-9/apoptosome activation in UV-induced apoptotic cells, the extrinsic apoptotic signaling generated by caspase-8 activation and consequent activation of B-cell lymphoma extra long (Bid) to release cytochrome c from mitochondria was observed. Thus, the post-mitochondrial apoptotic pathway downstream of cytochrome c release cannot operate the apoptosome function in UV-induced apoptosis in thymic 3SB cells. The intracellular redistribution and sequestration of apoptosis-related proteins upon mitochondrion-based apoptotic signaling was identified as a novel cellular mechanism to respond to DNA damage in an agent type-specific manner. This finding suggests that the kind of the critical ultimate apoptosis-inducing DNA lesion complex form resulting from the agent-specific DNA damage responses is important to determine which of apoptosis signals would be activated.


Subject(s)
Caspase 9/metabolism , Radiation, Ionizing , Thymus Gland/radiation effects , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Fractionation , Cells, Cultured , Cytochromes c/metabolism , Enzyme Activation/radiation effects , Mice , Mitochondria/metabolism , Thymus Gland/metabolism
8.
Mol Biol Cell ; 18(3): 1107-17, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17215513

ABSTRACT

Disassembly of the nucleolus during mitosis is driven by phosphorylation of nucleolar proteins. RNA processing stops until completion of nucleolar reformation in G(1) phase. Here, we describe the RNA methyltransferase NSUN2, a novel substrate of Aurora-B that contains an NOL1/NOP2/sun domain. NSUN2 was concentrated in the nucleolus during interphase and was distributed in the perichromosome and cytoplasm during mitosis. Aurora-B phosphorylated NSUN2 at Ser139. Nucleolar proteins NPM1/nucleophosmin/B23 and nucleolin/C23 were associated with NSUN2 during interphase. In mitotic cells, association between NPM1 and NSUN2 was inhibited, but NSUN2-S139A was constitutively associated with NPM1. The Aurora inhibitor Hesperadin induced association of NSUN2 with NPM1 even in mitosis, despite the silver staining nucleolar organizer region disassembly. In vitro methylation experiments revealed that the Aurora-B-phosphorylation and the phosphorylation-mimic mutation (S139E) suppressed methyltransferase activities of NSUN2. These results indicate that Aurora-B participates to regulate the assembly of nucleolar RNA-processing machinery and the RNA methyltransferase activity of NSUN2 via phosphorylation at Ser139 during mitosis.


Subject(s)
Methyltransferases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Amino Acid Sequence , Animals , Aurora Kinase B , Aurora Kinases , Cell Nucleolus/drug effects , Cell Nucleolus/enzymology , Conserved Sequence , HeLa Cells , Hesperidin/pharmacology , Histones/metabolism , Humans , Interphase/drug effects , Methyltransferases/chemistry , Mitosis/drug effects , Models, Biological , Molecular Sequence Data , Molecular Weight , Nuclear Proteins/metabolism , Nucleophosmin , Phosphoproteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Transport/drug effects , RNA-Binding Proteins/metabolism , Vertebrates , Nucleolin
9.
Dev Cell ; 4(4): 549-60, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12689593

ABSTRACT

Cell division is finely controlled by various molecules including small G proteins and kinases/phosphatases. Among these, Aurora B, RhoA, and the GAP MgcRacGAP have been implicated in cytokinesis, but their underlying mechanisms of action have remained unclear. Here, we show that MgcRacGAP colocalizes with Aurora B and RhoA, but not Rac1/Cdc42, at the midbody. We also report that Aurora B phosphorylates MgcRacGAP on serine residues and that this modification induces latent GAP activity toward RhoA in vitro. Expression of a kinase-defective mutant of Aurora B disrupts cytokinesis and inhibits phosphorylation of MgcRacGAP at Ser387, but not its localization to the midbody. Overexpression of a phosphorylation-deficient MgcRacGAP-S387A mutant, but not phosphorylation-mimic MgcRacGAP-S387D mutant, arrests cytokinesis at a late stage and induces polyploidy. Together, these findings indicate that during cytokinesis, MgcRacGAP, previously known as a GAP for Rac/Cdc42, is functionally converted to a RhoGAP through phosphorylation by Aurora B.


Subject(s)
Cell Division/genetics , Eukaryotic Cells/enzymology , GTPase-Activating Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Aurora Kinase B , Aurora Kinases , Contractile Proteins/genetics , Contractile Proteins/metabolism , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Enzymologic/genetics , HeLa Cells , Humans , Immunohistochemistry , Mitosis/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Transport/genetics , Serine/genetics , Serine/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
10.
Mol Carcinog ; 48(9): 810-20, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19204928

ABSTRACT

Aurora-A, also known as Aik, BTAK, or STK15, is a centrosomal serine/threonine protein kinase, which is proto-oncogenic and is overexpressed in a wide range of human cancers. Besides gene amplification and mRNA overexpression, proteolytic resistance mechanisms are thought to contribute to overexpression of Aurora-A. However, it is not yet clear how overexpressed Aurora-A affects the expression of transformed phenotype. Here, we found that nuclear accumulation of Aurora-A was critical for transformation activity. Cellular protein fractionation experiments and immunoblot analysis demonstrated a predominance of Aurora-A in the nuclear soluble fraction in head and neck cancer cells. Indirect immunofluorescence using confocal laser microscopy confirmed nuclear Aurora-A in head and neck cancer cells, while most oral keratinocytes exhibited only centrosomal localization. The expression of nuclear export signal-fused Aurora-A demonstrated that the oncogenic transformation activity was lost on disruption of the nuclear localization. Thus, the cytoplasmic localization of overexpressed Aurora-A previously demonstrated by immunohistochemical analysis is not likely to correspond to that in intact cancer cells. This study identifies an alternative mode of Aurora-A overexpression in cancer, through nuclear rather than cytoplasmic functions. We suggest that substrates of Aurora-A in the cell nuclear soluble fraction can represent a novel therapeutic target for cancer.


Subject(s)
Cell Nucleus/metabolism , Cell Transformation, Neoplastic , Protein Serine-Threonine Kinases/metabolism , Animals , Aurora Kinase A , Aurora Kinases , BALB 3T3 Cells , Cell Line , Cell Line, Tumor , Centrosome/metabolism , Cytoplasm/metabolism , Fluorescent Antibody Technique, Indirect , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunoblotting , Immunoprecipitation , Keratinocytes/cytology , Keratinocytes/metabolism , Mice , Microscopy, Confocal , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
11.
Virchows Arch ; 450(3): 297-302, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17235564

ABSTRACT

Aurora-B kinase is a chromosomal passenger protein and is essential for chromosome segregation and cytokinesis. Aurora-B overexpression in various cancer cells induces chromosomal number instability to produce multinuclearity and relates to metastasis. Here, we examined the expression of Aurora-B in oral squamous cell carcinoma (OSCC) to elucidate the relationship between Aurora-B expression and clinico-pathological findings by immunohistochemistry. Aurora-B expression was observed in normal oral squamous epithelia and OSCC cases, but the number of positive cells was significantly higher in OSCC than in normal squamous epithelium (p < 0.01). The labeling index of Aurora-B was significantly correlated with lymph node metastasis (p < 0.01) and histological grades of differentiation (p < 0.01). We also compared Aurora-B expression with Ki-67 expression and a positive correlation was found (p < 0.0001). Moreover, Aurora-B expression is significantly more frequent in multinuclear tumor cells than in total tumor cells. In summary, we found that Aurora-B expression was well correlated with cell proliferation, induction of multinuclear cells, histological differentiation, and metastasis in OSCC. These findings suggest that Aurora-B may be involved in tumor progression and that Aurora-B can be a new diagnostic and therapeutic target for OSCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Adult , Aged , Aged, 80 and over , Aurora Kinase B , Aurora Kinases , Carcinoma, Squamous Cell/secondary , Cell Count , Cell Proliferation , Epithelium/metabolism , Epithelium/pathology , Female , Humans , Immunoenzyme Techniques , Ki-67 Antigen/metabolism , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Middle Aged , Mouth Neoplasms/pathology , Neoplasm Metastasis
12.
Mol Biol Cell ; 13(4): 1099-108, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11950924

ABSTRACT

Aurora B is a protein kinase and a chromosomal passenger protein that undergoes dynamic redistribution during mitosis. We have probed the mechanism that regulates its localization with cells expressing green fluorescent protein (GFP)-tagged wild-type or mutant aurora B. Aurora B was found at centromeres at prophase and persisted until approximately 0.5 min after anaphase onset, when it redistributed to the spindle midzone and became concentrated at the equator along midzone microtubules. Depolymerization of microtubules inhibited the dissociation of aurora B from centromeres at early anaphase and caused the dispersion of aurora B from the spindle midzone at late anaphase; however, centromeric association during prometaphase was unaffected. Inhibition of CDK1 deactivation similarly caused aurora B to remain associated with centromeres during anaphase. In contrast, inhibition of the kinase activity of aurora B appeared to have no effect on its interactions with centromeres or initial relocation onto midzone microtubules. Instead, kinase-inactive aurora B caused abnormal mitosis and deactivation of the spindle checkpoint. In addition, midzone microtubule bundles became destabilized and aurora B dispersed from the equator. Our results suggest that microtubules, CDK1, and the kinase activity each play a distinct role in the dynamics and functions of aurora B in dividing cells.


Subject(s)
Cell Division , Mitosis , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Animals , Aurora Kinase B , Aurora Kinases , Cells, Cultured , Cyclin B/metabolism , Green Fluorescent Proteins , Kidney/metabolism , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Microtubules/metabolism , Models, Biological , Plasmids/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Rhodamines/metabolism , Time Factors , Transcription, Genetic , Transfection , Tubulin/metabolism
13.
Mol Biol Cell ; 14(4): 1489-500, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12686604

ABSTRACT

Aurora-B is a protein kinase required for chromosome segregation and the progression of cytokinesis during the cell cycle. We report here that Aurora-B phosphorylates GFAP and desmin in vitro, and this phosphorylation leads to a reduction in filament forming ability. The sites phosphorylated by Aurora-B; Thr-7/Ser-13/Ser-38 of GFAP, and Thr-16 of desmin are common with those related to Rho-associated kinase (Rho-kinase), which has been reported to phosphorylate GFAP and desmin at cleavage furrow during cytokinesis. We identified Ser-59 of desmin to be a specific site phosphorylated by Aurora-B in vitro. Use of an antibody that specifically recognized desmin phosphorylated at Ser-59 led to the finding that the site is also phosphorylated specifically at the cleavage furrow during cytokinesis in Saos-2 cells. Desmin mutants, in which in vitro phosphorylation sites by Aurora-B and/or Rho-kinase are changed to Ala or Gly, cause dramatic defects in filament separation between daughter cells in cytokinesis. The results presented here suggest the possibility that Aurora-B may regulate cleavage furrow-specific phosphorylation and segregation of type III IFs coordinatedly with Rho-kinase during cytokinesis.


Subject(s)
Desmin/chemistry , Desmin/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Aurora Kinase B , Aurora Kinases , Binding Sites/genetics , COS Cells , Cell Division , Cell Line , Desmin/genetics , Glial Fibrillary Acidic Protein/chemistry , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , In Vitro Techniques , Intermediate Filaments/metabolism , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine/chemistry , rho-Associated Kinases
14.
Mol Biol Cell ; 14(1): 78-92, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12529428

ABSTRACT

Survivin, a member of the inhibitor of apoptosis protein family, has attracted growing attention due to its expression in various tumors and its potential application in tumor therapy. However, its subcellular localization and function have remained controversial: Recent studies revealed that survivin is localized at the mitotic spindle, binds caspases, and could thus protect cells from apoptosis. The cell cycle-dependent expression of survivin and its antiapoptotic function led to the hypothesis that survivin connects the cell cycle with apoptosis, thus providing a death switch for the termination of defective mitosis. In other studies, survivin was detected at kinetochores, cleavage furrow, and midbody, localizations being characteristic for chromosomal passenger proteins. These proteins are involved in cytokinesis as inferred from the observation that RNA interference and expression of mutant proteins led to cytokinesis defects without an increase in apoptosis. To remedy these discrepancies, we analyzed the localizations of a survivinDsRed fusion protein in HeLa cells by using confocal laser scanning microscopy and time-lapse video imaging. SurvivinDsRed was excluded from the interphase nucleus and was detected in centrosomes and at kinetochores. It dissociated from chromosomes at the anaphase/telophase transition and accumulated at the ends of polar microtubuli where it was immediately condensed to the midbody. Overexpression of both survivinDsRed and of a phosphorylation-defective mutant conferred resistance against apoptosis-inducing reagents, but only the overexpressed mutant protein caused an aberrant cytokinesis. These data characterize in detail the dynamics of survivin in vertebrate cells and confirm that survivin represents a chromosomal passenger protein.


Subject(s)
Luminescent Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Apoptosis/physiology , Cell Division/physiology , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Kinetochores/metabolism , Luminescent Proteins/genetics , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Mutation , Neoplasm Proteins , Recombinant Fusion Proteins/genetics , Survivin , Red Fluorescent Protein
15.
Mol Med Rep ; 15(4): 1722-1726, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28260067

ABSTRACT

Rho GDP-dissociation inhibitor Ɵ (RhoGDIƟ), a regulator of the Rho family of proteins, is expressed abundantly in the hematopoietic cell lineage. During apoptosis of hematopoietic cells, RhoGDIƟ is cleaved by caspaseĀ­3 at Asp19 and this cleaved form (Δ19Ā­RhoGDIƟ) has been implicated in the apoptotic pathway. To clarify the role of RhoGDIƟ in hematopoietic cells, the present study performed immunoblotting and immunofluorescence staining to examine the expression of RhoGDIƟ and ∆19Ā­RhoGDIƟ during phorbol 12Ā­myristate 13Ā­acetate (PMA)Ā­stimulated differentiation of human THPĀ­1 monocytic cells to macrophages. During differentiation of the THPĀ­1 cells to macrophages, the expression of RhoGDIƟ remained stable; however, the expression of Δ19Ā­RhoGDIƟ increased, particularly in wellĀ­spreading, nonĀ­apoptotic cells, which differentiated into macrophages. These results suggested that Δ19Ā­RhoGDIƟ has an apoptosisĀ­independent role in the PMAĀ­induced differentiation of THPĀ­1 cells to macrophages.


Subject(s)
Apoptosis/drug effects , Aspartic Acid/metabolism , Cell Differentiation/drug effects , Macrophages/cytology , Macrophages/metabolism , Tetradecanoylphorbol Acetate/pharmacology , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , Cell Line , Humans , Macrophages/drug effects
16.
Oncol Lett ; 14(5): 5688-5694, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29113196

ABSTRACT

Bovine lactoferrin (bLF) is a multifunctional protein with anti-inflammatory, antibacterial, antiviral, anti-tumour and immunoregulatory effects. The present study was conducted to evaluate the anti-inflammatory and anti-tumour effects of liposomal bLF (LbLF) in a 1,2-dimethylhydrazine (DMH)/dextran sulphate sodium (DSS)-induced model of carcinogenesis in F344 rats. F344 rats were randomly divided into three groups: Control (water), 500 or 1,000 mg/kg/day LbLF; additionally, the rats were injected with DMH (20 mg/kg) once per week for 8 consecutive weeks, after one week of drinking water containing 1% DSS. All rats were sacrificed at 25 weeks. The tissues were examined for the presence of aberrant crypt foci (ACF) and subjected to histopathological analysis. Additionally, human colon cancer cells were utilised to investigate the effect of LbLF on proliferation and inflammation. Rats from the 500 and 1,000 mg/kg/day LbLF groups harboured significantly fewer colon ACF, adenomas and adenocarcinomas than the rats from the control group. Lastly, it was demonstrated that LbLF inhibits cell growth and TNF-α mRNA expression. These data support the hypothesis that LbLF affects colorectal carcinogenesis by suppressing inflammation and cell proliferation in rats.

17.
Oncogene ; 24(49): 7266-72, 2005 Nov 10.
Article in English | MEDLINE | ID: mdl-16027732

ABSTRACT

Aurora-B, previously known as AIM-1, is a conserved eukaryotic mitotic protein kinase. In mammals, this kinase plays an essential role in chromosomal segregation processes, including chromosome condensation, alignment, control of spindle checkpoints, chromosome segregation, and cytokinesis. Aurora-B is overexpressed in various cancer cells, suggesting that the kinase activity perturbs chromosomal segregation processes. Its forced overexpression induces chromosomal number instability and progressive tumorigenicity in rodent cells in vitro and in vivo. Nevertheless, based on focus formation in BALB/c 3T3 A31-1-1 cells, Aurora-B is not oncogenic. Here, we show that Aurora-B kinase activity augments Ras-mediated cell transformation. RNA interference with short hairpin RNA inhibits transformation by Ras and its upstream oncogene Src, but not by the downstream oncogene Raf. In addition, the inner centromere protein, which is a passenger protein associated with Aurora-B, has a similar ability to potentiate the activity of oncogenic Ras. These data indicate that elevated Aurora-B activity promotes transformation by oncogenic Ras by enhancing oncogenic signaling and by converting chromosome number-stable cells to aneuploid cells.


Subject(s)
Aneuploidy , Cell Transformation, Neoplastic/metabolism , Genes, ras/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Aurora Kinase B , Aurora Kinases , BALB 3T3 Cells/cytology , BALB 3T3 Cells/drug effects , BALB 3T3 Cells/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Genes, src/physiology , Mice , Nocodazole/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Interference , raf Kinases/physiology
18.
Oncogene ; 24(6): 1122-7, 2005 Feb 03.
Article in English | MEDLINE | ID: mdl-15592510

ABSTRACT

Aurora kinases are known to play a key role in maintaining mitotic fidelity, and overexpression of aurora kinases has been noted in various tumors. Overexpression of aurora kinase activity is thought to promote cancer development through a loss of centrosome or chromosome number integrity. Here we observed augmentation of G12V-mutated HRAS-induced neoplastic transformation in BALB/c 3T3 A31-1-1 cells transfected with Aurora-A. Aurora-A-short hairpin RNA (shRNA) experiments showed that the expression level of Aurora-A determines susceptibility to transformation. Aurora-A gene amplification was noted in human patients with tongue or gingival squamous carcinoma (4/11). Amplification was observed even in pathologically normal epithelial tissue taken at sites distant from the tumors in two patients with tongue cancer. However, overexpression of Aurora-A mRNA was observed only within the tumors of all patients examined (11/11). Our data indicate that Aurora-A gene amplification and overexpression play a role in human carcinogenesis, largely due to the effect of Aurora-A on oncogenic cell growth, rather than a loss of maintenance of centrosomal or chromosomal integrity.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/physiopathology , Cell Transformation, Neoplastic/genetics , Gene Amplification , Gene Expression Regulation, Neoplastic , Gingival Neoplasms/genetics , Gingival Neoplasms/physiopathology , Protein Kinases/biosynthesis , Tongue Neoplasms/genetics , Tongue Neoplasms/physiopathology , Aurora Kinases , Base Sequence , Cell Cycle Proteins , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Molecular Sequence Data , Polymorphism, Genetic , Protein Serine-Threonine Kinases , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Xenopus Proteins , ras Proteins/genetics , ras Proteins/pharmacology
19.
Clin Exp Metastasis ; 23(7-8): 323-34, 2006.
Article in English | MEDLINE | ID: mdl-17111235

ABSTRACT

Rho guanine nucleotide dissociation inhibitors (RhoGDIs) regulate the activity of Rho family GTPases. RhoGDIbeta (LyGDI/GDID4/RhoGDI2) has two caspase cleavage sites after Asp19 and Asp55. The resulting cleavage products, DeltaN(1-19)RhoGDIbeta and DeltaN(1-55)RhoGDIbeta, are expressed in cells under conditions that activate caspases. DeltaN(1-19)RhoGDIbeta, which can inhibit GDP dissociation, is implicated in the process of apoptosis, whereas the physiological roles for DeltaN(1-55)RhoGDIbeta, which lacks the ability to inhibit GDP dissociation, are largely unknown. To explore the roles of DeltaN(1-55)RhoGDIbeta, we examined the phenotypes of v-src-transformed metastatic fibroblasts transfected with plasmids for expressing DeltaN(1-55)RhoGDIbeta. Although the expression of DeltaN(1-55)RhoGDIbeta had no effect on the rate of growth in vitro, it suppressed experimental metastasis and decreased the rate of growth in vivo. In addition, DeltaN(1-55)RhoGDIbeta-expressing cells had enhanced adhesion to fibronectin, laminin, and collagens but reduced retention in the lung after intravenous injection. Also, the expression of DeltaN(1-55)RhoGDIbeta promoted anoikis without affecting the levels of activated Rac1 or Cdc42. Furthermore, DeltaN(1-55)RhoGDIbeta did not affect the expression or phosphorylation of focal adhesion kinase, p44/p42 mitogen-activated protein kinases, or Akt1 before or after induction of anoikis. Thus, DeltaN(1-55)RhoGDIbeta appears to promote anoikis by undefined mechanisms, thereby suppressing metastasis in v-src-transformed fibroblasts.


Subject(s)
Anoikis , Guanine Nucleotide Dissociation Inhibitors/physiology , Neoplasm Metastasis/prevention & control , Animals , Cell Transformation, Neoplastic , Genes, src , Guanine Nucleotide Dissociation Inhibitors/chemistry , Male , Mice , Mice, Inbred BALB C , Minor Histocompatibility Antigens , Signal Transduction , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors
20.
Cancer Res ; 62(18): 5168-77, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12234980

ABSTRACT

Phosphorylation of histone H3 at Ser-10 is required for maintenance of properchromosome dynamics during mitosis. AIM-1, a mammalian Ipl1/aurora kinase involved in H3 phosphorylation, is transcriptionally overexpressed in many tumor cell lines. Increased expression of the AIM-1 gene has been observed in human colorectal tumors of advanced grade and stage. Here we report that forced exogenous overexpression of AIM-1 in Chinese hamster embryo cells causes increased mitotic Ser-10 phosphorylation with concomitant induction of lagging chromosomes during mitosis. Lagging chromosomes could also be induced by transfection with mutated histone H3 (S10E), which is thought to maintain Ser-10 in the phosphorylated state. In the present study, chromosome number instability and increased tumor invasiveness were noted in constitutively AIM-1-overexpressing cells in vivo. Increased mitotic Ser-10 phosphorylation was also observed in various colorectal tumor cells with high AIM-1 expression levels. These data suggest that increased H3 histone phosphorylation as a result of AIM-1 overexpression is a major precipitating factor of chromosome instability and, thus, may play a role in carcinogenesis.


Subject(s)
Aneuploidy , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Histones/metabolism , Protein Kinases/biosynthesis , Protein Serine-Threonine Kinases , Animals , Aurora Kinase B , Aurora Kinases , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cricetinae , Cricetulus , Fibroblasts/physiology , Mice , Mice, Inbred BALB C , Mice, Nude , Mitosis/genetics , Mitosis/physiology , Phosphorylation , Protein Kinases/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL