Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Int. microbiol ; 19(3): 133-141, sept. 2016. ilus, tab, graf
Article in English | IBECS | ID: ibc-162890

ABSTRACT

Phosphorus is a pivotal element in all biochemical systems: it serves to store metabolic energy as ATP, it forms the backbone of genetic material such as RNA and DNA, and it separates cells from the environment as phospholipids. In addition to this 'big hits', phosphorus has recently been shown to play an important role in other important processes such as cell cycle regulation. In the present review, we briefly summarize the biological processes in which phosphorus is involved in the yeast Saccharomyces cerevisiae before discussing our latest findings on the role of this element in the regulation of DNA replication in this eukaryotic model organism. We describe both the role of phosphorus in the regulation of G1 progression by means of the Cyclin Dependent Kinase (CDK) Pho85 and the stabilization of the cyclin Cln3, as well as the role of other molecule composed of phosphorus-the polyphosphate-in cell cycle progression, dNTP synthesis, and genome stability. Given the eminent role played by phosphorus in life, we outline the future of phosphorus in the context of one of the main challenges in human health: cancer treatment (AU)


No disponible


Subject(s)
Saccharomyces cerevisiae/growth & development , Phosphorus/physiology , Cyclin-Dependent Kinases/physiology , Antineoplastic Agents/pharmacology , Polyphosphates/analysis , Eukaryotic Cells/physiology , Cell Cycle/physiology , Cell Cycle Proteins/analysis
2.
Acta Pharmacol Sin ; 30(4): 442-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19305423

ABSTRACT

AIM: The aim of this study was to investigate the mechanism of pseudolaric acid B (PLAB)-induced cell cycle arrest in human melanoma SK-28 cells. METHODS: Cell growth inhibition was detected by MTT assay, the cell cycle was analyzed by flow cytometry, and protein expression was examined by Western blot analysis. RESULTS: PLAB inhibited the growth of human melanoma cells and induced G(2)/M arrest in SK-28 cells, accompanied by an up-regulation of Cdc2 phosphorylation and a subsequent down-regulation of Cdc2 expression. Furthermore, PLAB decreased the expression of Cdc25C phosphatase and increased the expression of Wee1 kinase. Meanwhile, a reduction in Cdc2 activity was partly due to induction of the expression of p21(waf1/cip1) in a p53-dependent manner. In addition, PLAB activated the checkpoint kinase, Chk2, and increased the expression of p53, two major targets of ATM kinase. These effects were inhibited by caffeine, an ATM kinase inhibitor. We also found that PLAB significantly enhanced ATM kinase activity. CONCLUSION: Taken together, these results suggest that PLAB induced G(2)/M arrest in human melanoma cells via a mechanism involving the activation of ATM, and the effect of PLAB on Cdc2 activity was mediated via interactions with the Chk2-Cdc25C and p53 signalling pathways, two distinct downstream pathways of ATM. PLAB may be a promising chemopreventive agent for treating human melanoma.


Subject(s)
Cell Cycle Proteins/physiology , Cell Division/drug effects , DNA-Binding Proteins/physiology , Diterpenes/pharmacology , Drugs, Chinese Herbal/pharmacology , G2 Phase/drug effects , Protein Serine-Threonine Kinases/physiology , Signal Transduction/drug effects , Tumor Suppressor Proteins/physiology , Ataxia Telangiectasia Mutated Proteins , CDC2 Protein Kinase , Caffeine/pharmacology , Cell Cycle Proteins/analysis , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 2 , Cyclin B/analysis , Cyclin B1 , Cyclin-Dependent Kinase Inhibitor p21/analysis , Cyclin-Dependent Kinases , Humans , Melanoma/pathology , Nuclear Proteins/analysis , Protein-Tyrosine Kinases/analysis , Tumor Suppressor Protein p53/analysis , cdc25 Phosphatases/analysis
3.
Article in English | MEDLINE | ID: mdl-18540058

ABSTRACT

The initiation of eukaryotic DNA replication requires the tightly controlled assembly of a set of replication factors. Mcm10 is a highly conserved nuclear protein that plays a key role in the initiation and elongation processes of DNA replication by providing a physical link between the Mcm2-7 complex and DNA polymerases. The central domain, which contains the CCCH zinc-binding motif, is most conserved within Mcm10 and binds to DNA and several proteins, including proliferative cell nuclear antigen. In this study, the central domain of human Mcm10 was crystallized using the hanging-drop vapour-diffusion method in the presence of PEG 3350. An X-ray diffraction data set was collected to a resolution of 2.6 A on a synchrotron beamline. The crystals formed belonged to space group R3, with unit-cell parameters a = b = 99.5, c = 133.0 A. According to Matthews coefficient calculations, the crystals were predicted to contain six MCM10 central domain molecules in the asymmetric unit.


Subject(s)
Cell Cycle Proteins/analysis , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/isolation & purification , Amino Acid Motifs , Amino Acid Sequence , Cell Cycle Proteins/genetics , Cell Line , Cloning, Molecular , DNA, Complementary , Escherichia coli/genetics , Gene Library , Genetic Vectors , Humans , Kidney/cytology , Minichromosome Maintenance Proteins , Molecular Sequence Data , Nucleic Acid Amplification Techniques , Protein Structure, Tertiary , X-Ray Diffraction
4.
Phytother Res ; 22(3): 416-23, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18058993

ABSTRACT

Although verticinone, a major alkaloid isolated from the bulbus of Fritillaria ussuriensis, has been shown to induce differentiation in human leukemia cells, the exact mechanism of this action is not completely understood in cancer cells. Verticinone was used to conduct growth and apoptosis-related experiments for two stages of oral cancer on immortalized human oral keratinocytes (IHOKs) and primary oral cancer cells (HN4). The procedures included MTT assay, three-dimensional (3-D) raft cultures, Western blotting, cell cycle analysis, nuclear staining and cytochrome c expression related to the apoptosis signaling pathway. Verticinone inhibited the proliferation of immortalized and malignant oral keratinocytes in a dose- and time-dependent manner. In 3-D organotypic culture, verticinone-treated cells were less mature than the control cells, displaying low surface keratinization and decreased epithelial thickness. The major mechanism by which verticinone inhibits growth appears to be induced apoptosis and G(0)G(1) cell cycle arrest. This finding is supported by the results of the cell cycle analysis, FITC-Annexin V staining, DNA fragmentation assay and Hoechst 33258 staining. Furthermore, the cytosolic level of cytochrome c was increased, while the expression of Bcl-2 protein was gradually down-regulated and Bax was up-regulated, accompanied by caspase-3 activation. The data suggests that verticinone may induce apoptosis through a caspase pathway mediated by mitochondrial damage in immortalized keratinocytes and oral cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cevanes/pharmacology , Keratinocytes/drug effects , Antibodies/metabolism , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/biosynthesis , Carcinoma/pathology , Cell Cycle Proteins/analysis , Cell Cycle Proteins/biosynthesis , Cell Line, Transformed , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/pharmacology , Fritillaria/chemistry , Gene Expression/drug effects , Humans , Mouth Neoplasms/pathology , Time Factors
5.
Oncogene ; 25(7): 1053-69, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16205633

ABSTRACT

Here, we assessed and compared the anticancer efficacy and associated mechanisms of silymarin and silibinin in human prostate cancer (PCA) PC3 cells; silymarin is comprised of silibinin and its other stereoisomers, including isosilybin A, isosilybin B, silydianin, silychristin and isosilychristin. Silymarin and silibinin (50-100 microg/ml) inhibited cell proliferation, induced cell death, and caused G1 and G2-M cell cycle arrest in a dose/time-dependent manner. Molecular studies showed that G1 arrest was associated with a decrease in cyclin D1, cyclin D3, cyclin E, cyclin-dependent kinase (CDK)4, CDK6 and CDK2 protein levels, and CDK2 and CDK4 kinase activity, together with an increase in CDK inhibitors (CDKIs) Kip1/p27 and Cip1/p21. Further, both agents caused cytoplasmic sequestration of cyclin D1 and CDK2, contributing to G1 arrest. The G2-M arrest by silibinin and silymarin was associated with decreased levels of cyclin B1, cyclin A, pCdc2 (Tyr15), Cdc2, and an inhibition of Cdc2 kinase activity. Both agents also decreased the levels of Cdc25B and cell division cycle 25C (Cdc25C) phosphatases with an increased phosphorylation of Cdc25C at Ser216 and its translocation from nucleus to the cytoplasm, which was accompanied by an increased binding with 14-3-3beta. Both agents also increased checkpoint kinase (Chk)2 phosphorylation at Thr68 and Ser19 sites, which is known to phosphorylate Cdc25C at Ser216 site. Chk2-specific small interfering RNA largely attenuated the silymarin and silibinin-induced G2-M arrest. An increase in the phosphorylation of histone 2AX and ataxia telangiectasia mutated was also observed. These findings indicate that silymarin and silibinin modulate G1 phase cyclins-CDKs-CDKIs for G1 arrest, and the Chk2-Cdc25C-Cdc2/cyclin B1 pathway for G2-M arrest, together with an altered subcellular localization of critical cell cycle regulators. Overall, we observed comparable effects for both silymarin and silibinin at equal concentrations by weight, suggesting that silibinin could be a major cell cycle-inhibitory component in silymarin. However, other silibinin stereoisomers present in silymarin also contribute to its efficacy, and could be of interest for future investigation.


Subject(s)
Carcinoma/drug therapy , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinases/metabolism , Flavones/therapeutic use , Prostatic Neoplasms/drug therapy , Silymarin/therapeutic use , Cell Cycle/drug effects , Cell Cycle Proteins/analysis , Cell Nucleus/chemistry , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/analysis , Cytoplasm/chemistry , Humans , Male , Phosphorylation , Silybin , Tumor Cells, Cultured
6.
Zhong Xi Yi Jie He Xue Bao ; 2(2): 120-2, 2004 Mar.
Article in Chinese | MEDLINE | ID: mdl-15339473

ABSTRACT

OBJECTIVE: To investigate the role of p27 in the inhibition of emodin on the mesangial cell (MC) proliferation induced by tumor necrosis factor-alpha(TNF-alpha). METHODS: p27 protein of MC was detected with western blotting analysis. The degree of MC proliferation was estimated through [(3)H] thymidine ([(3)H] TdR) incorporation. Different dosage of emodin (50 mg/L,100 mg/L) was added into MC stimulated by TNF-alpha. RESULTS: TNF-alpha (200 kU/L) decreased p27 level of MC cultured in serum-free DMEM for 24 hours and increased[(3)H] TdR incorporation. Emodin increased p27 level of MC stimulated by TNF-alpha and decreased [(3)H] TdR incorporation. The more the emodin was added, the greater the above-mentioned effect of emodin. CONCLUSION: The increment of p27 level maybe play an important role in the inhibition of emodin on MC proliferation induced by TNF-alpha.


Subject(s)
Cell Cycle Proteins/physiology , Emodin/antagonists & inhibitors , Glomerular Mesangium/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/physiology , Animals , Cell Cycle Proteins/analysis , Cell Division/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27 , Glomerular Mesangium/cytology , Rats , Rats, Sprague-Dawley , Tumor Suppressor Proteins/analysis
7.
Gene Expr ; 11(5-6): 271-8, 2004.
Article in English | MEDLINE | ID: mdl-15200239

ABSTRACT

Septin 3 is a novel member of the septin subfamily of GTPase domain proteins. Human septin 3 was originally cloned during a screening of genes expressed in human teratocarcinoma cells induced to differentiate with retinoic acid. Alternative splicing of the septin 3 gene transcript produces two isoforms, A and B, in the human brain, though their regional expression and physiological function remain to be determined. The purpose of the present study was to identify the expression patterns of human septin 3 isoforms in normal human brain and a human neuroblastoma cell line, SH-SY5Y, after retinoic acid-induced differentiation. The expression and distribution patterns of septin 3 isoforms A and B were similar and resembled that of another septin, CDCrel-1. Septin 3A and 3B were expressed in normal human brain in a region-specific manner, with the highest level in the temporal cortex and hippocampus and the lowest level in the brainstem regions. Prominent immunoreactivity was observed diffusely in the neocortices in association with neuropils and punctate structures suggestive of synaptic junctions. Immunoprecipitation studies revealed that septin 3A, 3B, and CDCrel-1 form a complex in the frontal cortex of human brain. These findings, taken together, suggest that septin 3A and 3B, along with CDCrel-1, play some fundamental role(s) in synaptogenesis and neuronal development.


Subject(s)
Brain/metabolism , GTP Phosphohydrolases/metabolism , Antibodies/immunology , Brain Chemistry , Cell Cycle Proteins/analysis , Cell Differentiation , Cell Line , Frontal Lobe/chemistry , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/immunology , Gene Expression , Humans , Immunochemistry , Immunoprecipitation , Protein Binding , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/analysis , Septins , Tretinoin/pharmacology , Up-Regulation
8.
J Neurosurg ; 98(2): 378-84, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12593626

ABSTRACT

OBJECT: Ionizing radiation is the gold-standard adjuvant treatment for glioblastoma multiforme (GBM), the most aggressive primary brain tumor. The mechanisms underlying neoplastic glial cell growth inhibition after administration of ionizing radiation, however, remain largely unknown. In this report, the authors characterize the response of GBM cells to ionizing radiation and elucidate factors that correlate with the radiosensitivity of these tumors. METHODS: Six human GBM cell lines were subjected to increasing doses of radiation. Each demonstrated a dose-dependent suppression of cell proliferation. In the most radiosensitive cell line, the authors demonstrated a transient increase in the expression of the cyclin-dependent kinase inhibitors (CDKIs) p21 and p27, which corresponded with a G1 cell-cycle arrest. In contrast, the most radioresistant cell line demonstrated a decrease in p21 and p27 expression levels, which correlated with a failure to arrest. Apoptosis did not occur in any cell line following irradiation. Instead, autophagic cell changes were observed following administration of radiation, regardless of the relative radiosensitivity of the cell line. CONCLUSIONS: These findings elucidate some of the molecular responses of GBMs to irradiation and suggest novel targets for future therapy.


Subject(s)
Autophagy/genetics , Autophagy/radiation effects , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Cycle Proteins/radiation effects , Cyclins/radiation effects , Enzyme Inhibitors/radiation effects , G1 Phase/genetics , G1 Phase/radiation effects , Glioblastoma/pathology , Glioblastoma/radiotherapy , Radiation, Ionizing , Tumor Suppressor Proteins/radiation effects , Brain Neoplasms/genetics , Cell Cycle Proteins/analysis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/analysis , Enzyme Inhibitors/analysis , Glioblastoma/genetics , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Organelles/genetics , Organelles/radiation effects , Radiation Tolerance/genetics , Radiation Tolerance/radiation effects , Time Factors , Tumor Cells, Cultured/pathology , Tumor Cells, Cultured/radiation effects , Tumor Suppressor Proteins/analysis
9.
J Inorg Biochem ; 88(1): 25-36, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11750022

ABSTRACT

The reaction of [Cu(dien)NO(3)]NO(3) with 2-amino-5-methylthiazole (2A5MT), 2-amino-2-thiazoline (2A-2Tzn), imidazole (im), N,N'-thiocarbonyldiimidazole (Tcdim), 2-aminothiazole (2AT) and 2-ethylimidazole (2Etim), gave a new series of mixed-ligand compounds of the general formula [Cu(dien)(B)NO(3))]NO(3); (dien, diethylenetriamine; B, 2A5MT, 2A-2Tzn, im, Tcdim, 2AT and 2Etim). The complexes have been characterised by elemental analysis, molar conductivity and magnetic measurements, as well as by electronic and IR spectral studies. According to the above measurements the possible structure of the compounds is the square pyramidal in the solid state and the square planar in aqueous solution. We tested all complexes for antiproliferative (cytostatic and cytotoxic) activity against a panel of cell lines (HeLa, L929, HT-29 and T47D). All [(dien)Cu(B)NO(3))](NO(3)) complexes had an activity against colon cancer cells (HT-29), inducing G2/M cell cycle arrest, an effect that for most of the complexes could be attributed to p34cdc2 inhibition by tyrosine-phosphorylation and/or to induction of (cyclin-dependent kinase inhibitor) p21(WAF1). Other cell lines were resistant to the majority of the complexes, except [Cu(dien)(2A5MT)NO(3))](NO(3)), that had showed the highest anti-proliferative activity against HT-29 cells also. The predilection for colon cancer cells and the relatively low toxicity against normal (L929) cells justify further investigation of this group of compounds.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Copper/chemistry , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Animals , Antineoplastic Agents/toxicity , Cell Cycle/drug effects , Cell Cycle Proteins/analysis , Cell Division/drug effects , Colonic Neoplasms/pathology , Copper/toxicity , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Imidazoles/toxicity , Ligands , Molecular Structure , Organometallic Compounds/toxicity , Polyamines/chemistry , Spectrum Analysis , Structure-Activity Relationship , Thiazoles/chemical synthesis , Thiazoles/pharmacology , Thiazoles/toxicity , Tumor Cells, Cultured/drug effects
10.
Dev Neurosci ; 23(2): 100-6, 2001.
Article in English | MEDLINE | ID: mdl-11509832

ABSTRACT

Previously we have shown that changes in maternal dietary choline are associated with permanent behavioral changes in offspring. Importantly, in adult male rats, feeding a choline-deficient diet increases the localization of cyclin-dependent kinase inhibitors (CDKIs) in the liver, whereas young adult CDKI knockout mice (p15Ink4B or p27Kip1) exhibit behavioral abnormalities. Thus, maternal dietary choline-CDKI interactions could underlie the changes we observe in fetal hippocampal development and cognitive function in offspring. Here, timed-pregnant rats on embryonic day E12 were fed the AIN-76 diet with varying levels of dietary choline for 6 days, and, on E18, fetal brain sections were collected, and the localization of CDKI proteins was studied using immunohistochemistry and an unbiased image analysis method. In choline-supplemented animals compared to controls, the number of cells with nuclear immunoreactivity for p15Ink4b CDKI protein was decreased 2- to 3-fold in neuroepithelial ventricular zones and adjacent subventricular zones corresponding to the fimbria, primordial dentate gyrus and Ammon's horn regions in the fetal hippocampus. In contrast, maternal dietary choline deficiency significantly decreased nuclear p15Ink4b immunoreactivity in the neuroepithelial layer of the dentate gyrus. Unlike p15Ink4b, the CDKI protein p27Kip1 was observed almost exclusively in the cytoplasm, though the protein was distributed throughout the proliferating and postmitotic zones in the E18 fetal hippocampus. Maternal dietary choline supplementation decreased the cytoplasmic staining intensity for p27Kip1 throughout the fetal hippocampus compared to control animals. Choline deficiency increased the staining intensity of p27Kip1 throughout the hippocampus in association with increased expression of MAP-1 and vimentin proteins. These results link maternal dietary choline availability to CDKI protein immunoreactivity and commitment to differentiation during fetal hippocampal development.


Subject(s)
Cell Cycle Proteins/analysis , Choline/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/analysis , Dentate Gyrus/chemistry , Dentate Gyrus/embryology , Tumor Suppressor Proteins/analysis , Animal Nutritional Physiological Phenomena , Animals , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p27 , Female , Memory , Microtubule-Associated Proteins/analysis , Pregnancy , Rats , Rats, Sprague-Dawley , Vimentin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL