Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 224
Filter
1.
Dev Biol ; 434(1): 84-95, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29198563

ABSTRACT

Transcriptional silencing is a conserved process used by embryonic germ cells to repress somatic fate and maintain totipotency and immortality. In Drosophila, this transcriptional silencing is mediated by polar granule component (pgc). Here, we show that in the adult ovary, pgc is required for timely germline stem cell (GSC) differentiation. Pgc is expressed transiently in the immediate GSC daughter (pre-cystoblast), where it mediates a pulse of transcriptional silencing. This transcriptional silencing mediated by pgc indirectly promotes the accumulation of Cyclin B (CycB) and cell cycle progression into late-G2 phase, when the differentiation factor bag of marbles (bam) is expressed. Pgc mediated accumulation of CycB is also required for heterochromatin deposition, which protects the germ line genome against selfish DNA elements. Our results suggest that transient transcriptional silencing in the pre-cystoblast "re-programs" it away from self-renewal and toward the gamete differentiation program.


Subject(s)
Cell Differentiation/physiology , G2 Phase/physiology , Gene Silencing/physiology , Germ Cells/metabolism , Stem Cells/metabolism , Animals , Cyclin B/biosynthesis , Cyclin B/genetics , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Germ Cells/cytology , Heterochromatin/genetics , Heterochromatin/metabolism , Stem Cells/cytology
2.
Mol Reprod Dev ; 83(12): 1070-1082, 2016 12.
Article in English | MEDLINE | ID: mdl-27699901

ABSTRACT

Sea urchin eggs exhibit a cap-dependent increase in protein synthesis within minutes after fertilization. This rise in protein synthesis occurs at a constant rate for a great number of proteins translated from the different available mRNAs. Surprisingly, we found that cyclin B, a major cell-cycle regulator, follows a synthesis pattern that is distinct from the global protein population, so we developed a mathematical model to analyze this dissimilarity in biosynthesis kinetic patterns. The model includes two pathways for cyclin B mRNA entry into the translational machinery: one from immediately available mRNA (mRNAcyclinB) and one from mRNA activated solely after fertilization (XXmRNAcyclinB). Two coefficients, α and ß, were added to fit the measured scales of global protein and cyclin B synthesis, respectively. The model was simplified to identify the synthesis parameters and to allow its simulation. The calculated parameters for activation of the specific cyclin B synthesis pathway after fertilization included a kinetic constant (ka ) of 0.024 sec-1 , for the activation of XXmRNAcyclinB, and a critical time interval (t2 ) of 42 min. The proportion of XXmRNAcyclinB form was also calculated to be largely dominant over the mRNAcyclinB form. Regulation of cyclin B biosynthesis is an example of a select protein whose translation is controlled by pathways that are distinct from housekeeping proteins, even though both involve the same cap-dependent initiation pathway. Therefore, this model should help provide insight to the signaling utilized for the biosynthesis of cyclin B and other select proteins. Mol. Reprod. Dev. 83: 1070-1082, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cyclin B/biosynthesis , Fertilization , Models, Biological , Ovum/metabolism , Protein Biosynthesis/physiology , RNA, Messenger, Stored/metabolism , Animals , Female , Ovum/cytology , Sea Urchins/metabolism
3.
Gene ; 594(1): 82-88, 2016 Dec 05.
Article in English | MEDLINE | ID: mdl-27601256

ABSTRACT

The sea cucumber, Apostichopus japonicus, is a good model for studying environmentally-induced aestivation by a marine invertebrate. One of the central requirements of aestivation is the repression of energy-expensive cellular processes such as cell cycle progression. The present study identified the gene structure of the cell cycle regulator, cyclin B, and detected the expression levels of this gene over three stages of the annual aestivation-arousal cycle. Furthermore, the DNA methylation characteristics of cyclin B were analyzed in non-aestivation and deep-aestivation stages of sea cucumbers. We found that the cyclin B promoter contains a CpG island, three CCAAT-boxes and three cell cycle gene homology regions (CHRs). Application of qRT-PCR analysis showed significant downregulation of cyclin B transcript levels during deep-aestivation in comparison with non-aestivation in both intestine and longitudinal muscle, and these returned to basal levels after arousal from aestivation. Methylation analysis of the cyclin B core promoter revealed that its methylation level showed significant differences between non-aestivation and deep-aestivation stages (p<0.05) and interestingly, a positive correlation between Cyclin B transcripts expression and methylation levels of the core promoter was also observed. Our findings suggest that cell cycle progression may be reversibly arrested during aestivation as indicated by the changes in cyclin B expression levels and we propose that DNA methylation is one of the regulatory mechanisms involved in cyclin B transcriptional variation.


Subject(s)
Cyclin B , DNA Methylation/physiology , Estivation/physiology , Gene Expression Regulation/physiology , Promoter Regions, Genetic/physiology , Sea Cucumbers , Animals , Cyclin B/biosynthesis , Cyclin B/genetics , Sea Cucumbers/genetics , Sea Cucumbers/metabolism
4.
J Pharm Pharmacol ; 68(7): 901-11, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27138740

ABSTRACT

OBJECTIVES: Our study aimed to evaluate the effect of cordycepin on human lung cancer cell lines A549 and NCI-H460. METHODS: Human lung cancer A549 cells and NCI-H460 cells were treated with different concentrations of cordycepin for different times. Cells incubated without cordycepin were defined as a control. The cell proliferation, migration and apoptosis were, respectively, determined by MTT assay, transwell migration assay and flow cytometry. Additionally, the expression levels of related proteins associated with cell cycle, epithelial-mesenchymal transition (EMT) and apoptosis were examined. KEY FINDINGS: The survival rate of A549 cells and NCI-H460 cells treated with cordycepin significantly decreased compared with untreated cells in a concentration-dependent manner, while the apoptosis rate increased. The migration number of cells treated with cordycepin significantly decreased as the increase in concentration. qRT-PCR and Western blot analysis showed that the aberrant expression of related molecules associated with cell cycle, migration and apoptosis was observed in the lung cancer cells, such as cyclin B, cyclin E, MMP-9, caspase-3 and Bcl-2. CONCLUSIONS: Cordycepin may exert inhibitory effects on the development of human lung cancer via inhibiting cell proliferation, suppressing migration and inducing apoptosis, suggesting that cordycepin may have therapeutic potential for the treatment of this disease.


Subject(s)
Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Deoxyadenosines/pharmacology , Cadherins/biosynthesis , Caspase 3/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Cyclin B/biosynthesis , Cyclin D/biosynthesis , Cyclin E/biosynthesis , Dose-Response Relationship, Drug , Humans , Matrix Metalloproteinase 9/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Time Factors , Vimentin/biosynthesis
5.
Fungal Genet Biol ; 91: 6-19, 2016 06.
Article in English | MEDLINE | ID: mdl-26994663

ABSTRACT

In Saccharomyces cerevisiae, the S-phase cyclin Clb6 is expressed shortly before the G1/S transition. It has been shown that in S phase the SCF(Cdc4) ubiquitin ligase controls Clb6 proteolysis, which requires cyclin-dependent kinases activity. A Clb6-3A mutant, bearing non-phosphorylatable mutations at S6A, T39A, and S147A, was observed to be hyperstabilized in S-phase but was unstable in mitosis. In this study, we found that the APC(Cdh1) form of the Anaphase-Promoting Complex (APC) was required for Clb6 proteolysis in both early and late G1. An in vitro ubiquitination assay confirmed that Clb6 is a substrate for APC(Cdh1). A KEN box and a destruction box in the Clb6N-terminus were identified. Mutations in the KEN box (mkb) and/or the destruction box (mdb) enhanced Clb6 stability in G1. Expression of Clb6mkd, bearing both mutations in the mkb and mdb, allowed cells to bypass the late G1 arrest caused by cdc4-1. This bypass phenotype was observed to depend upon CDK phosphorylation at residues S6, T39 and S147. Compared to Clb6, overexpression of Clb6ST, bearing all five mutations of S6A, T39A, S147A, mkb and mdb in combination, had a greater effect on promoting expression of Clb2 and S-phase entry, caused a greater G2 delay and a greater defect in cell division. Swe1 was also required for bud emergence when Clb6ST was overexpressed. Our observations suggest that both APC(Cdh1) and SCF(Cdc4)-dependent proteolysis of Clb6 at the G1/S border are crucial for multiple cell cycle regulated events including proper expression of Clb2, the G1/S and G2/M cell cycle transitions and for proper completion of cell division at mitotic exit.


Subject(s)
Cdh1 Proteins/genetics , Cell Cycle Proteins/genetics , Cyclin B/genetics , F-Box Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Anaphase-Promoting Complex-Cyclosome/genetics , Cell Cycle/genetics , Cyclin B/biosynthesis , Gene Expression Regulation, Fungal , Mitosis , Mutation , Phosphorylation , Protein-Tyrosine Kinases/genetics , Proteolysis , S Phase/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/biosynthesis
6.
PLoS One ; 11(3): e0150318, 2016.
Article in English | MEDLINE | ID: mdl-26962866

ABSTRACT

The cyclin B/CDK1 complex is a key regulator of mitotic entry. Using PP242, a specific ATP-competitive inhibitor of mTOR kinase, we provide evidence that the mTOR signalling pathway controls cyclin B mRNA translation following fertilization in Sphaerechinus granularis and Paracentrotus lividus. We show that PP242 inhibits the degradation of the cap-dependent translation repressor 4E-BP (eukaryotic initiation factor 4E-Binding Protein). PP242 inhibits global protein synthesis, delays cyclin B accumulation, cyclin B/CDK1 complex activation and consequently entry into the mitotic phase of the cell cycle triggered by fertilization. PP242 inhibits cyclin B mRNA recruitment into active polysomes triggered by fertilization. An amount of cyclin B mRNA present in active polysomes appears to be insensitive to PP242 treatment. Taken together, our results suggest that, following sea urchin egg fertilization, cyclin B mRNA translation is controlled by two independent mechanisms: a PP242-sensitive and an additional PP242-insentitive mechanism.


Subject(s)
Cyclin B/biosynthesis , Embryo, Nonmammalian/metabolism , Fertilization/physiology , Protein Biosynthesis/physiology , TOR Serine-Threonine Kinases/metabolism , Animals , Fertilization/drug effects , Indoles/pharmacology , Peptide Initiation Factors/metabolism , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Purines/pharmacology , RNA, Messenger/metabolism , Sea Urchins/metabolism
7.
Biochim Biophys Acta ; 1859(4): 627-38, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26921500

ABSTRACT

The heterotrimeric CCAAT-binding factor NF-Y controls the expression of a multitude of genes involved in cell cycle progression. NF-YA is present in two alternatively spliced isoforms, NF-YAs and NF-YAl, differing in 28 aminoacids in the N-terminal Q-rich activation domain. NF-YAs has been identified as a regulator of stemness and proliferation in mouse embryonic cells (mESCs) and human hematopoietic stem cells (hHSCs), whereas the role of NF-YAl is not clear. In the muscle system, NF-YA expression is observed in proliferating cells, but barely detectable in terminally differentiated cells in vitro and adult skeletal muscle in vivo. Here, we show that NF-YA inactivation in mouse myoblasts impairs both proliferation and differentiation. The overexpression of the two NF-YA isoforms differentially affects myoblasts fate: NF-YAs enhance cell proliferation, while NF-YAl boosts differentiation. The molecular mechanisms were investigated by expression profilings, detailing the opposite programs of the two isoforms. Bioinformatic analysis of the regulated promoters failed to detect a significant presence of CCAAT boxes in the regulated genes. NF-YAl activates directly Mef2D, Six genes, and p57kip2 (Cdkn1c), and indirectly the myogenic regulatory factors (MRFs). Specifically, Cdkn1c activation is induced by NF-Y binding to its CCAAT promoter and by reducing the expression of the lncRNA Kcnq1ot1, a negative regulator of Cdkn1c transcription. Overall, our results indicate that NF-YA alternative splicing is an influential muscle cell determinant, through direct regulation of selected cell cycle blocking genes, and, directly and indirectly, of muscle-specific transcription factors.


Subject(s)
CCAAT-Binding Factor/genetics , Cell Differentiation/genetics , Muscle Development/genetics , Muscle, Skeletal/growth & development , Protein Isoforms/genetics , Animals , CCAAT-Binding Factor/biosynthesis , Cell Proliferation/genetics , Cyclin B/biosynthesis , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Humans , Mice , Myoblasts/metabolism , Promoter Regions, Genetic , Transcription Factors/biosynthesis , Transcription Factors/genetics
8.
BMC Cancer ; 16: 17, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26762417

ABSTRACT

BACKGROUND: Silvestrol is a cyclopenta[b]benzofuran that was isolated from the fruits and twigs of Aglaia foveolata, a plant indigenous to Borneo in Southeast Asia. The purpose of the current study was to determine if inhibition of protein synthesis caused by silvestrol triggers autophagy and apoptosis in cultured human cancer cells derived from solid tumors. METHODS: In vitro cell viability, flow cytometry, fluorescence microscopy, qPCR and immunoblot was used to study the mechanism of action of silvestrol in MDA-MB-435 melanoma cells. RESULTS: By 24 h, a decrease in cyclin B and cyclin D expression was observed in silvestrol-treated cells relative to control. In addition, silvestrol blocked progression through the cell cycle at the G2-phase. In silvestrol-treated cells, DAPI staining of nuclear chromatin displayed nucleosomal fragments. Annexin V staining demonstrated an increase in apoptotic cells after silvestrol treatment. Silvestrol induced caspase-3 activation and apoptotic cell death in a time- and dose-dependent manner. Furthermore, both silvestrol and SAHA enhanced autophagosome formation in MDA-MB-435 cells. MDA-MB-435 cells responded to silvestrol treatment with accumulation of LC3-II and time-dependent p62 degradation. Bafilomycin A, an autophagy inhibitor, resulted in the accumulation of LC3 in cells treated with silvestrol. Silvestrol-mediated cell death was attenuated in ATG7-null mouse embryonic fibroblasts (MEFs) lacking a functional autophagy protein. CONCLUSIONS: Silvestrol potently inhibits cell growth and induces cell death in human melanoma cells through induction of early autophagy and caspase-mediated apoptosis. Silvestrol represents a natural product scaffold that exhibits potent cytotoxic activity and could be used for the further study of autophagy and its relationship to apoptosis in cancer cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Caspase 3/biosynthesis , Melanoma/drug therapy , Triterpenes/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Caspase 3/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B/biosynthesis , Cyclin D/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Macrolides/administration & dosage , Melanoma/genetics , Melanoma/pathology , Mice
9.
Int J Mol Med ; 37(2): 526-32, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26709398

ABSTRACT

Although amygdalin is used by many cancer patients as an antitumor agent, there is a lack of information on the efficacy and toxicity of this natural compound. In the present study, the inhibitory effect of amygdalin on the growth of renal cell carcinoma (RCC) cells was examined. Amygdalin (10 mg/ml) was applied to the RCC cell lines, Caki-1, KTC-26 and A498, for 24 h or 2 weeks. Untreated cells served as controls. Tumor cell growth and proliferation were determined using MTT and BrdU tests, and cell cycle phases were evaluated. Expression of the cell cycle activating proteins cdk1, cdk2, cdk4, cyclin A, cyclin B, cyclin D1 and D3 as well as of the cell cycle inhibiting proteins p19 and p27 was examined by western blot analysis. Surface expression of the differentiation markers E- and N-cadherin was also investigated. Functional blockade by siRNA was used to determine the impact of several proteins on tumor cell growth. Amygdalin treatment caused a significant reduction in RCC cell growth and proliferation. This effect was correlated with a reduced percentage of G2/M-phase RCC cells and an increased percentage of cells in the G0/1-phase (Caki-1 and A498) or cell cycle arrest in the S-phase (KTC-26). Furthermore, amygdalin induced a marked decrease in cell cycle activating proteins, in particular cdk1 and cyclin B. Functional blocking of cdk1 and cyclin B resulted in significantly diminished tumor cell growth in all three RCC cell lines. Aside from its inhibitory effects on growth, amygdalin also modulated the differentiation markers, E- and N-cadherin. Hence, exposing RCC cells to amygdalin inhibited cell cycle progression and tumor cell growth by impairing cdk1 and cyclin B expression. Moreover, we noted that amygdalin affected differentiation markers. Thus, we suggest that amygdalin exerted RCC antitumor effects in vitro.


Subject(s)
Amygdalin/administration & dosage , Carcinoma, Renal Cell/drug therapy , Cyclin B/biosynthesis , Cyclin-Dependent Kinases/biosynthesis , Apoptosis/drug effects , CDC2 Protein Kinase , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B/genetics , Cyclin-Dependent Kinases/genetics , Gene Expression Regulation, Neoplastic , Humans
10.
Oncol Rep ; 34(6): 3288-96, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26503118

ABSTRACT

Acute myeloid leukemia (AML) is a disorder involving hematopoietic stem cells, characterized by blockage of hematopoietic cell differentiation and an increase in clonal neoplastic proliferation. AML is associated with poor patient outcome. PBK/TOPK is a protein kinase derived from PDZ-binding kinase (PBK)/T-lymphokine-activated killer (T-LAK) cell-originated protein kinase (TOPK). Previous studies have shown that PBK/TOPK is expressed in hematologic tumors. In the present study, we aimed to investigate the role of PBK/TOPK in promyelocyte proliferation and to clarify the molecular mechanism. PBK/TOPK knockdown (KD) significantly decreased cell proliferation and viability in the NB4 and HL-60 promyelocytes. PBK/TOPK KD resulted in G2/M cell cycle arrest that attributed to a decrease in cdc2 and cyclin B expression. In addition, PBK/TOPK KD caused apoptosis, as evidenced by activation of the mitochondrial apoptotic pathway and an increase in TUNEL-positive cells. PBK/TOPK KD induced mitochondrial dysfunction and ROS generation, and inhibition of mitochondrial dysfunction and ROS production suppressed PBK/TOPK KD-induced cell cycle arrest and apoptosis. Moreover, PBK/TOPK KD decreased Nrf2 expression and ARE-binding activity. Overexpression of Nrf2 inhibited the PBK/TOPK KD-induced decrease in cdc2 and cyclin B expression and cell cycle arrest, and blocked ROS production and apoptosis. Based on literature and our results, it was demonstrated that Nrf2 may be a crucial regulator that mediates PBK/TOPK-exerted promotion of cell proliferation. PBK/TOPK stabilizes Nrf2, strictly regulates the ROS level, promotes cell cycle progression and inhibits apoptosis, contributing to the proliferation of promyelocytes. Our results provide new insights into the molecular mechanism of PBK/TOPK-mediated promyelocyte proliferation and shed light on the pathogenesis of AML.


Subject(s)
Cell Proliferation/genetics , Leukemia, Myeloid, Acute/genetics , Mitogen-Activated Protein Kinase Kinases/biosynthesis , NF-E2-Related Factor 2/genetics , Apoptosis/genetics , CDC2 Protein Kinase , Cell Cycle Checkpoints/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cyclin B/biosynthesis , Cyclin-Dependent Kinases/biosynthesis , Gene Expression Regulation, Leukemic , Granulocyte Precursor Cells/metabolism , Granulocyte Precursor Cells/pathology , HL-60 Cells , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Mitogen-Activated Protein Kinase Kinases/genetics , NF-E2-Related Factor 2/biosynthesis , Reactive Oxygen Species/metabolism
11.
Reproduction ; 150(4): 343-55, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26159832

ABSTRACT

Previously, we demonstrated the expression of resistin in the porcine ovary, the regulation of its expression and its direct effect on ovarian steroidogenesis. The objective of this study was to examine the effect of resistin on cell proliferation and apoptosis in a co-culture model of porcine granulosa and theca cells. First, we analysed the effect of resistin at 1 and 10  ng/ml alone or in combination with FSH- and IGF1 on ovarian cell proliferation with an alamarBlue assay and protein expression of cyclins A and B using western blot. Next, the mRNA and protein expression of selected pro-apoptotic and pro-survival regulators of cell apoptosis, caspase-9, -8 and -3 activity and DNA fragmentation using real time PCR, western blot, fluorescent assay and an ELISA kit, respectively, were analysed after resistin treatment. Furthermore, we determined the effect of resistin on the protein expression of ERK1/2, Stat and Akt kinase. Using specific inhibitors of these kinases, we also checked caspase-3 activity and protein expression. We found that resistin, at both doses, has no effect on cell proliferation. The results showed that resistin decreased pro-apoptotic genes, which was confirmed on protein expression of selected factors. We demonstrate an inhibitory effect of resistin on caspase activity and DNA fragmentation. Finally, resistin stimulated phosphorylation of the ERK1/2, Stat and Akt and kinases inhibitors reversed resistin action on caspase-3 activity and protein expression to control. All of these results showed that resistin has an inhibitory effect on porcine ovarian cell apoptosis by activation of the MAPK/ERK, JAK/Stat and Akt/PI3 kinase signalling pathways.


Subject(s)
Ovarian Follicle/drug effects , Ovary/drug effects , Resistin/pharmacology , Animals , Apoptosis/drug effects , Caspases/biosynthesis , Cell Proliferation/drug effects , Cyclin A/biosynthesis , Cyclin B/biosynthesis , Female , Granulosa Cells/drug effects , MAP Kinase Signaling System/drug effects , Oncogene Protein v-akt/drug effects , Ovarian Follicle/cytology , Ovary/cytology , Pregnancy , Swine , Theca Cells/drug effects
12.
J Ovarian Res ; 8: 35, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-26055813

ABSTRACT

BACKGROUND: Ovarian cancer represents the most fatal type of gynecological malignancies. Unfortunately, there are still no effective targeted treatment strategies for ovarian cancer. Overexpression of CRM1 has been correlated with poor prognosis of patients with ovarian cancer. AIM: In this study, we investigated the antitumor effects of a novel reversible inhibitor of CRM1 in ovarian cancer cells. METHODS: The effects of S109 on proliferation was detected by CCK-8, EdU, clonogenic assay. The protein expression were determined by Western blot. The subcellular localization of RanBP1 was analyzed by immunofluorescence microscopy assay. RESULTS: We demonstrated that S109 could induce nuclear accumulation of RanBP1, a canonical biomarker for CRM1 inhibition. This effect was clearly reversible in the majority of the cells, whereas the inhibitory effect of LMB could not be reversed. Our data reveal that treatment with S109 results in decrease in proliferation and colonogenic capacity of ovarian cancer cells by arresting cell cycle. Mechanistically, S109 treatment increase the expression of the cyclin-dependent kinase inhibitor p21, while it reduced the expression of cell cycle promoting proteins, Cyclin D1 and Cyclin B. CRM1 level itself was also down-regulated following S109 treatment. Furthermore, the nuclei of cells incubated with S109 accumulated tumor suppressor proteins (Foxo1, p27 and IκB-α). More importantly, Cys528 mutation of CRM1 abolished the ability of S109 to block proliferation of ovarian cancer cells. CONCLUSIONS: Together, our study identifies CRM1 as a valid target in ovarian cancer and provides a basis for the development of S109 in ovarian cancer.


Subject(s)
Aminopyridines/administration & dosage , Cyclopentanes/administration & dosage , Karyopherins/biosynthesis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B/biosynthesis , Cyclin D1 , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Karyopherins/antagonists & inhibitors , Ovarian Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Exportin 1 Protein
13.
Tumour Biol ; 36(2): 953-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25315186

ABSTRACT

Cyclins are a group of cell cycle regulatory proteins. Cyclin B acts as an activator to cyclin-dependent kinase 1 (CDK1), a protein kinase essential for G2/M phase transition. Deregulation of cyclins has been linked to a number of malignant neoplasms, but the impact on clinicopathological parameters seems to be cancer-specific. Overexpression of cyclin B has been shown to affect survival in some malignant tumors, including breast and esophageal cancer, but its impact on endometrial cancer has not been extensively studied. For this study, 211 endometrial endometrioid adenocarcinoma samples were obtained from patients surgically treated at the Oulu University Hospital. The samples were immunohistochemically stained and analyzed for cyclin B expression. The relationships between cyclin B expression and conventional prognostic factors were analyzed. A discrimination threshold for survival analyses was calculated by utilizing the receiver operator characteristic (ROC) method. Cyclin B expression correlated with grade and advanced stage. Survival analyses showed that cyclin B expression affects cancer-specific survival in univariate analysis. In multivariate analysis, the results were indicative that cyclin B may hold independent prognostic significance, but further studies are required to assess this.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Endometrioid/genetics , Cyclin B/biosynthesis , Prognosis , Adult , Aged , Aged, 80 and over , CDC2 Protein Kinase , Carcinoma, Endometrioid/pathology , Cyclin-Dependent Kinases/genetics , Female , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Staging , Survival Analysis
14.
J Huazhong Univ Sci Technolog Med Sci ; 34(2): 157-160, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24710924

ABSTRACT

Autophagy dysregulation, mitochondrial dynamic abnormality and cell cycle re-entry are implicated in the vulnerable neurons of patients with Alzheimer's disease. This study was designed to testify the association among autophagy, mitochondrial dynamics and cell cycle in dividing neuroblastoma (N2a) cells. The N2a cells were cultured in vitro and treated with different concentrations of 3-methyladenine (3-MA). The cell viability was detected by methyl thiazolyl tetrazolium (MTT) assay. They were randomly divided into control group (cells cultured in normal culture medium) and 3-MA group (cells treated with 10 mmol/L 3-MA). The cell cycle was analyzed in the two groups 3, 6, 12, and 24 h after treatment by flow cytometry. Western blotting was used to evaluate the expression levels of mitofission 1 (Fis1), mitofusin 2 (Mfn2), microtubule-associated protein 1 light chain 3 (LC3), cell cycle-dependent kinase 4 (CDK4) and cdc2. The flow cytometry revealed that the proportion of cells in G(2)/M was significantly increased, and that in G0/G1 was significantly reduced in the 3-MA group as compared with the control group. Western blotting showed that the expression levels of Fis1, LC3, and CDK4 were significantly up-regulated in the 3-MA group at the four indicated time points as compared with the control group. Mfn2 was initially decreased in the 3-MA group, and then significantly increased at 6 h or 12 h. Cdc2 was significantly increased in the 3-MA group at 3 h and 6 h, and then dropped significantly at 12 h and 24 h. Our data indicated that 3-MA-induced suppressed autophagy may interfere with the cell cycle progression and mitochondrial dynamics, and cause cell death. There are interactions among cell cycle, mitochondrial dynamics and autophagy in neurons.


Subject(s)
Autophagy/drug effects , Cell Cycle/drug effects , Adenine/administration & dosage , Adenine/analogs & derivatives , Apoptosis/drug effects , Autophagy/genetics , CDC2 Protein Kinase , Cell Cycle/genetics , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin B/biosynthesis , Cyclin-Dependent Kinases , Gene Expression Regulation/drug effects , Humans , Membrane Proteins/biosynthesis , Microtubule-Associated Proteins/biosynthesis , Mitochondrial Dynamics/drug effects , Mitochondrial Dynamics/genetics , Mitochondrial Proteins/biosynthesis , Neuroblastoma , Signal Transduction/drug effects
15.
Oncol Rep ; 31(1): 183-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24247637

ABSTRACT

Despite the fact that paclitaxel and doxorubicin are widely used as chemotherapy agents against several types of cancer, their combined effects on esophageal squamous cell carcinoma (ESCC) have never been fully elucidated. The present study was designed to investigate the biological effects of paclitaxel and doxorubicin in ESCC cells. Combination treatment with paclitaxel and doxorubicin significantly inhibited the proliferation of TE-12 cells in a dose-and time-dependent manner compared to treatment with paclitaxel or doxorubicin alone. FACS analysis showed that the percentage of cells in the G2/M phase was significantly increased at 12 h after treatment with the combination. Increased p-cdc2, p-Wee1 and p53 protein levels were observed, while Akt activation was suppressed by combination treatment with paclitaxel and doxorubicin. In addition, treatment with paclitaxel plus doxorubicin significantly increased apoptosis as indicated by increased cleaved poly(ADP-ribose) polymerase and cleaved caspase-7 and -9 levels. These results suggest that combination treatment with paclitaxel and doxorubicin induced G2/M cell cycle arrest and apoptosis in human ESCC cells by suppressing Akt activity. These findings highlight the potent apoptotic effect of combination therapy with paclitaxel and doxorubicin in ESCC cells and the potential clinical benefits of these two drugs in esophageal cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Doxorubicin/therapeutic use , Esophageal Neoplasms/drug therapy , M Phase Cell Cycle Checkpoints/drug effects , Paclitaxel/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Apoptosis/drug effects , CDC2 Protein Kinase , Caspase 7/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin B/biosynthesis , Cyclin-Dependent Kinases , Esophageal Squamous Cell Carcinoma , Humans , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Tumor Suppressor Protein p53/biosynthesis
16.
PLoS One ; 8(9): e74379, 2013.
Article in English | MEDLINE | ID: mdl-24058555

ABSTRACT

Antimitotic agents are frequently used to treat solid tumors and hematologic malignancies. However, one major limitation of antimitotic approaches is mitotic slippage, which is driven by slow degradation of cyclin B during a mitotic block. The extent to which cyclin B levels decline is proposed to be governed by an equilibrium between cyclin B synthesis and degradation. It was recently shown that the 3' untranslated region (UTR) of the murine cyclin B mRNA contributes to the synthesis of cyclin B during mitosis in murine cells. Using a novel live-cell imaging-based technique allowing us to study synthesis and degradation of cyclin B simultaneously at the single cell level, we tested here the role of the human cyclin B 3'UTR in regulating cyclin B synthesis during mitosis in human cells. We observed that the cyclin B 3'UTR was not sufficient to enhance cyclin B synthesis in human U2Os, HeLa or hTERT RPE-1 cells. A better understanding of how the equilibrium of cyclin B is regulated in mitosis may contribute to the development of improved therapeutic approaches to prevent mitotic slippage in cancer cells treated with antimitotic agents.


Subject(s)
3' Untranslated Regions/genetics , Cyclin B/biosynthesis , Cyclin B/genetics , Mitosis/genetics , Animals , Cell Line , Chromosomes, Human/metabolism , Genes, Reporter , Humans , Kinetics , Mice , Microtubules/drug effects , Microtubules/metabolism , Mitosis/drug effects , Paclitaxel/pharmacology , Proteolysis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Toxicol Lett ; 217(3): 226-34, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23296099

ABSTRACT

Sterigmatocystin (ST), a common environmental contaminant found across the world, is generally recognized as a potential carcinogen, mutagen and teratogen. Our previous epidemiological studies suggested that ST exposure might be a risk factor for esophageal cancer. However, the direct effects of ST on human esophageal epithelial cells are currently unknown. In the present study, we examined the effect of treating a human esophageal epithelial cell line (Het-1A) with ST on DNA damage, DNA repair mechanisms, and cell cycle distribution. We found that ST treatment could induce DNA damage and lead to a G2 phase arrest, associated with a marked up-regulation of G2/M regulatory proteins, including Cyclin B1, Cdc2/p-Cdc2, and Cdc25C/p-Cdc25C. Additionally, we found that the expression of two mismatch repair (MMR) proteins, hMLH1 and hMSH2, was up-regulated at both the mRNA and protein levels after ST treatment, suggesting that ST could induce the MMR system in Het-1A cells. Interestingly, ST-induced G2 phase arrest was mediated by hMLH1 up-regulation, but was independent of hMSH2. Treatment with hMLH1-siRNA prevented the up-regulation of Cyclin B1, Cdc2/p-Cdc2 and Cdc25C/p-Cdc25C in ST-treated cells, thereby inhibiting the subsequent G2 phase arrest of Het-1A cells. Moreover, we found that hMLH1 may act as a direct sensor of ST-mediated DNA damage. In conclusion, the study demonstrated that ST caused DNA damage and triggered G2 phase arrest in Het-1A cells, and hMLH1 participated in the ST-induced G2 phase arrest by up-regulating G2/M regulatory proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , DNA Damage , Epithelial Cells/drug effects , Esophageal Neoplasms/chemically induced , G2 Phase Cell Cycle Checkpoints/drug effects , Nuclear Proteins/biosynthesis , Sterigmatocystin/toxicity , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Blotting, Western , CDC2 Protein Kinase , Cell Line , Comet Assay , Cyclin B/biosynthesis , Cyclin B/genetics , Cyclin B/metabolism , Cyclin B1/biosynthesis , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin-Dependent Kinases , DNA Mismatch Repair/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/genetics , Humans , MutL Protein Homolog 1 , MutL Proteins , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , cdc25 Phosphatases/biosynthesis , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
18.
Int J Mol Med ; 31(3): 726-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23314756

ABSTRACT

In the present study, in order to investigate the anticancer effects of O-desmethylangolensin (O-DMA) on human hepatocellular carcinoma Hep3B cells, we first examined the antiproliferative effect of O-DMA. When Hep3B cells were treated with O-DMA at various concentrations (5-200 µM) for 24, 48 or 72 h, cell proliferation decreased significantly in a dose- and time-dependent manner. Moreover, O-DMA exposure at the IC50 concentration for 72 h arrested cells at the G2/M phase, which was accompanied by a reduction in CDK1, and an increase in cyclin A and B. Under the same conditions, O-DMA significantly increased the number of sub-G1 phase cells. Additionally, an Annexin V assay revealed that exposure to O-DMA affected the rate of cell apoptosis. O-DMA caused the downregulation of Bcl-2 and upregulation of Bax, which led to cytochrome c release from the mitochondria and activation of caspase-3. Taken together, these data suggest that O-DMA exhibits anticancer activity by arresting the cell cycle at G2/M phase and causing mitochondrial-dependent apoptosis in Hep3B cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , G2 Phase Cell Cycle Checkpoints/drug effects , Isoflavones/pharmacology , Liver Neoplasms/drug therapy , CDC2 Protein Kinase/biosynthesis , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin A/biosynthesis , Cyclin B/biosynthesis , Cytochromes c/metabolism , Humans , Mitochondria/metabolism , Phytoestrogens/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis
19.
Mod Pathol ; 26(1): 87-94, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22918167

ABSTRACT

Assessment of proliferation is important in female breast cancer and individual treatment decisions are based upon its results, especially in the luminal subgroups. Gene expression analyses fail to group male breast cancer into the intrinsic subgroups previously established in female breast cancer. Even though proliferation has been shown to divide male breast cancer into molecular subgroups with different prognoses, the clinical importance of proliferation markers has not yet been elucidated. Previous studies in male breast cancer have demonstrated contradictory results regarding the prognostic impact of histological grade and Ki-67, parameters strongly associated with proliferation. The aim of the present project was to study proliferation in male breast cancer by assessing other proliferation-related markers viz. cyclins A, B, D1 and mitotic count. A total of 197 male breast cancer cases with accessible paraffin-embedded material and outcome data were investigated. Immunohistochemical stainings were performed on tissue microarrays. Kaplan-Meier estimates and the Cox proportional regression models were used for survival analyses with breast cancer death as the event. The subset of patients with high expression of cyclin A (hazard ratio (HR) 3.7; P=0.001) and B (HR 2.7; P=0.02) demonstrated a poorer survival. Furthermore, high mitotic count was associated with an increased risk of breast cancer death (HR 2.5; P=0.01). In contrast, cyclin D1 overexpression was predictive of better breast cancer survival (HR 0.3; P=0.001). In conclusion, high levels of cyclin A and B expression and an elevated mitotic count result in a two to threefold higher risk for breast cancer death, whereas cyclin D1 overexpression halves the risk. The clinical utility of these proliferation markers needs further elucidation.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms, Male/mortality , Breast Neoplasms, Male/pathology , Adult , Aged , Aged, 80 and over , Breast Neoplasms, Male/metabolism , Cell Proliferation , Cyclin A/analysis , Cyclin A/biosynthesis , Cyclin B/analysis , Cyclin B/biosynthesis , Cyclin D1/analysis , Cyclin D1/biosynthesis , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Mitotic Index , Neoplasm Grading , Neoplasm Staging , Tissue Array Analysis , Young Adult
20.
J Chemother ; 24(5): 268-78, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23182046

ABSTRACT

Novel quinuclidinone derivatives have been previously reported by our laboratory. In this study, we investigated the impact of two novel quinuclidinone derivatives 4 and 6 on apoptotic signaling in breast cancer cells (MCF-7) and their normal counterparts (MCF-12a). Our data revealed that derivatives 4 and 6 reduced proliferation and induced apoptosis in breast cancer cells. However, derivative 6 was less cytotoxic to normal breast epithelial cells than breast cancer cells; therefore, we focused on derivative 6 for further investigation. Flow cytometric analysis showed that quinuclidinone derivative 6 reduced the percentage of MCF-7 cells in G(2)/M which is confirmed by increased expression levels of cyclin B, while it arrests MCF12a in G1 phase judging from increased p21. Quinuclidinone derivative 6 increased expression levels of p53 and Bax at both protein and mRNA levels and reduced expression level of Mdm2, Bcl2, Akt and Bcl-XL It also increased mitochondrial apoptotic pathways by activating release of cytochrome c which is consistent with activation of caspase-9 as confirmed by caspase-9 inhibitor LEHD-CHO. Finally, it increased sphingomyelinase signaling and ceramide formation as well as its downstream targets ERK1/2, p38, and JNK. Inhibition of ERK1/2 with PD98059 exerted little effect on the derivative 6-induced apoptosis and p38 inhibition with SB203580 slightly lessened apoptosis, whereas inhibition of JNK with SP600125 markedly suppressed derivative 6-induced apoptosis. These results indicate that derivative-6 induced the activation of sphingomyelinase signaling and that JNK played a pivotal role in induction of apoptosis in human breast cancer cells. In vivo studies and molecular docking experiments are now in progress for further anticancer investigations.


Subject(s)
Breast Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Quinuclidines/pharmacology , Sphingomyelin Phosphodiesterase/metabolism , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Cycle/drug effects , Cell Line, Tumor , Ceramides/metabolism , Cyclin B/biosynthesis , Enzyme-Linked Immunosorbent Assay , Female , Humans , MAP Kinase Signaling System/physiology , Quinuclidines/therapeutic use , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sphingomyelin Phosphodiesterase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...