1,187 results on '"Resting Phase, Cell Cycle"'
Search Results
2. Cell Cycle Regulation in Macrophages and Susceptibility to HIV-1.
- Author
-
Ferreira IATM, Porterfield JZ, Gupta RK, and Mlcochova P
- Subjects
- Animals, Cell Cycle Checkpoints, Cells, Cultured, DNA Damage, G1 Phase, Gram-Negative Bacteria immunology, Histone Deacetylase Inhibitors pharmacology, Humans, Macrophages immunology, Phosphorylation, Resting Phase, Cell Cycle, SAM Domain and HD Domain-Containing Protein 1 metabolism, vif Gene Products, Human Immunodeficiency Virus metabolism, vpr Gene Products, Human Immunodeficiency Virus metabolism, Cell Cycle, HIV-1 physiology, Macrophages physiology, Macrophages virology
- Abstract
Macrophages are the first line of defence against invading pathogens. They play a crucial role in immunity but also in regeneration and homeostasis. Their remarkable plasticity in their phenotypes and function provides them with the ability to quickly respond to environmental changes and infection. Recent work shows that macrophages undergo cell cycle transition from a G0/terminally differentiated state to a G1 state. This G0-to-G1 transition presents a window of opportunity for HIV-1 infection. Macrophages are an important target for HIV-1 but express high levels of the deoxynucleotide-triphosphate hydrolase SAMHD1, which restricts viral DNA synthesis by decreasing levels of dNTPs. While the G0 state is non-permissive to HIV-1 infection, a G1 state is very permissive to HIV-1 infection. This is because macrophages in a G1 state switch off the antiviral restriction factor SAMHD1 by phosphorylation, thereby allowing productive HIV-1 infection. Here, we explore the macrophage cell cycle and the interplay between its regulation and permissivity to HIV-1 infection.
- Published
- 2020
- Full Text
- View/download PDF
3. Unraveling quiescence-specific repressive chromatin domains.
- Author
-
Swygert SG and Tsukiyama T
- Subjects
- Adenosine Triphosphatases metabolism, Chromatin chemistry, Chromatin Assembly and Disassembly genetics, DNA-Binding Proteins metabolism, Gene Expression Regulation, Genome-Wide Association Study, Multiprotein Complexes metabolism, Saccharomyces cerevisiae physiology, Saccharomyces cerevisiae Proteins chemistry, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae Proteins metabolism, Transcription, Genetic, Cell Cycle, Chromatin genetics, Chromatin metabolism, Resting Phase, Cell Cycle
- Abstract
Quiescence is a highly conserved inactive life stage in which the cell reversibly exits the cell cycle in response to external cues. Quiescence is essential for diverse processes such as the maintenance of adult stem cell stores, stress resistance, and longevity, and its misregulation has been implicated in cancer. Although the non-cycling nature of quiescent cells has made obtaining sufficient quantities of quiescent cells for study difficult, the development of a Saccharomyces cerevisiae model of quiescence has recently enabled detailed investigation into mechanisms underlying the quiescent state. Like their metazoan counterparts, quiescent budding yeast exhibit widespread transcriptional silencing and dramatic chromatin condensation. We have recently found that the structural maintenance of chromosomes (SMC) complex condensin binds throughout the quiescent budding yeast genome and induces the formation of large chromatin loop domains. In the absence of condensin, quiescent cell chromatin is decondensed and transcription is de-repressed. Here, we briefly discuss our findings in the larger context of the genome organization field.
- Published
- 2019
- Full Text
- View/download PDF
4. Newcastle disease virus induces G 0 /G 1 cell cycle arrest in asynchronously growing cells.
- Author
-
Wang Y, Wang R, Li Y, Sun Y, Song C, Zhan Y, Tan L, Liao Y, Meng C, Qiu X, and Ding C
- Subjects
- Cell Division genetics, Cell Proliferation, Cyclin D metabolism, DNA Replication, G1 Phase Cell Cycle Checkpoints, HeLa Cells, Host-Pathogen Interactions, Humans, Newcastle disease virus genetics, Signal Transduction, Transcription Factor CHOP metabolism, Cell Cycle, Cell Cycle Checkpoints, Newcastle disease virus physiology, Resting Phase, Cell Cycle, Virus Replication genetics
- Abstract
The cell cycle, as a basic cellular process, is conservatively regulated. Consequently, subversion of the host cell replication cycle is a common strategy employed by many viruses to create a cellular environment favorable for viral replication. Newcastle disease virus (NDV) causes disease in poultry and is also an effective oncolytic agent. However, the effects of NDV infection on cell cycle progression are unknown. In this study, we showed that NDV replication in asynchronized cells resulted in the accumulation of infected cells in the G
0 /G1 phase of the cell cycle, which benefitted the proliferation of NDV. Examination of various cell cycle-regulatory proteins showed that expression of cyclin D1, was significantly reduced following NDV infection. Importantly, the decreased expression of cyclin D1 was reversed by inhibition of CHOP expression, indicating that induction of the PERK-eIF-2a-ATF4-CHOP signaling pathway was involved in the G0 /G1 phase cell cycle arrest observed following NDV infection., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
5. Requirement of ClC-3 in G0/G1 to S Phase Transition Induced by IGF-1 via ERK1/2-Cyclins Cascade in Multiple Myeloma Cells.
- Author
-
Du Y, Tu YS, Tang YB, Huang YY, Zhou FM, Tian T, and Li XY
- Subjects
- Cell Cycle genetics, Cell Line, Tumor, Cells, Cultured, Chloride Channels genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cyclin-Dependent Kinase Inhibitor p27 genetics, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Cyclins genetics, Extracellular Signal-Regulated MAP Kinases genetics, G1 Phase, Humans, Multiple Myeloma genetics, Multiple Myeloma pathology, Resting Phase, Cell Cycle, S Phase, Tumor Cells, Cultured, Cell Cycle drug effects, Chloride Channels metabolism, Cyclins metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Insulin-Like Growth Factor I pharmacology, Multiple Myeloma metabolism
- Abstract
Background: ClC-3 is involved in the proliferation and migration of several cancer cells. However, ClC-3 expression and its role of cell-cycle control in multiple myeloma (MM) has not yet been investigated., Methods: MM cells were treated with different concentrations of IGF (30, 100, 300 ng/mL), and their proliferation was examined by CCK-8. The effects of ClC-3 on cell cycle progression was detected by flow cytometry. Western blot was used to analyze the relative levels of ClC3, CD138, P21, P27, CDK, p-Erk1/2, and t-Erk1/2 protein expression. Transfection of RPMI8226 with gpClC-3 cDNA and siRNA alters the expression of ClC-3., Results: We compared the expression of ClC-3 in primary myeloma cells and in MM cell lines (U266 and RPMI8266) with that in normal plasma cells (PCs) from normal subjects and found that myeloma cells from patients and MM cell lines had significantly higher expression of ClC-3. Additionally, silencing of ClC-3 with the small interfering RNA (siRNA) that targets human ClC-3 decreased proliferation of RPMI8226 after IGF-1 treatment and slowed cell cycle progression from G0/G1 to S phase, which was associated with diminished phosphorylation of ERK1/2, down-expression of cyclin E, cyclin D1 and up-regulation of p27 and p21. By contrast, overexpression of ClC-3 potentiated cell proliferation induced by IGF-1, raised the percentage of S phase cells, enhanced phosphorylation of ERK1/2, downregulated p27 and p21 and upregulated cyclin E and cyclin D1., Conclusions: ClC-3 accelerated G0/G1 to S phase transition in the cell cycle by modulating ERK1/2 kinase activity and expression of G1/S transition related proteins, making ClC-3 an attractive therapeutic target in MM.
- Published
- 2018
- Full Text
- View/download PDF
6. Cell cycle synchronization and analysis of apoptosis-related gene in skin fibroblasts from domestic cat (Felis silvestris catus) and kodkod (Leopardus guigna).
- Author
-
Veraguas D, Gallegos PF, Castro FO, and Rodriguez-Alvarez L
- Subjects
- Animals, Apoptosis genetics, Cell Survival, Cloning, Organism veterinary, Contact Inhibition, Culture Media, Serum-Free, Gene Expression Profiling, Resting Phase, Cell Cycle, Cell Cycle physiology, Felidae physiology, Fibroblasts cytology, Nuclear Transfer Techniques veterinary
- Abstract
The kodkod population is in constant decrease and the somatic cell nuclear transfer (SCNT) might help to preserve the genetic pool of this species. The cell cycle synchronization of donor cells plays a crucial role in SCNT. The objective of this research was to evaluate two different methods for quiescence induction, serum starvation (SS) and contact inhibition (CI), both for 1, 3 and 5 days, on skin fibroblast from domestic cat and kodkod. Flow cytometry analysis revealed that in domestic cat, SS and CI, both at 3 and 5 days, increased the percentage of fibroblasts in G0/G1 compared to growing cells (GC) (p < .05). In kodkod, only SS for 3 and 5 days and CI for 1 and 3 days increased the percentage of fibroblasts in G0/G1 compared to GC (p < .05). Viability analysis by differential staining revealed that SS for 5 days decreased the proportion of live fibroblasts in domestic cat and kodkod (p < .05). Regarding gene expression analysis, in domestic cat fibroblasts, no differences were found in the BAX/BCL2 ratio in SS and CI (both at 1, 3 and 5 days) compared to GC. In kodkod fibroblasts, BAX/BCL2 ratio was increased in CI at 3 and 5 days compared to SS at 3 and 5 days (p < .05). In conclusion, in kodkod fibroblasts SS for 5 days and CI after 3 days might have a negative impact on cellular viability. According to these results, we suggest SS for 3 days for cell cycle synchronization in kodkod fibroblasts., (© 2017 Blackwell Verlag GmbH.)
- Published
- 2017
- Full Text
- View/download PDF
7. Fast cell cycle analysis for intraoperative characterization of brain tumor margins and malignancy.
- Author
-
Alexiou GA, Vartholomatos G, Goussia A, Batistatou A, Tsamis K, Voulgaris S, and Kyritsis AP
- Subjects
- Adult, Aged, Aged, 80 and over, Brain Neoplasms diagnosis, DNA, Neoplasm analysis, DNA, Neoplasm genetics, Female, Flow Cytometry, G1 Phase, Glioblastoma pathology, Glioblastoma surgery, Glioma pathology, Glioma surgery, Humans, Intraoperative Care, Male, Meningioma pathology, Meningioma surgery, Middle Aged, Mitosis, ROC Curve, Resting Phase, Cell Cycle, S Phase, Young Adult, Brain Neoplasms pathology, Brain Neoplasms surgery, Cell Cycle
- Abstract
Flow cytometry, although indispensable for the characterization of hematologic malignancies, has not been extensively evaluated in solid tumors. To date intraoperative pathology evaluation of frozen sections of tissue obtained during surgery is the gold standard for intraoperative diagnosis. We investigated the value of a modified rapid protocol for cell cycle analysis for the intraoperative characterization of intracranial lesions and their surgical margins. We investigated patients who underwent surgery for an intracranial lesion suspicious for a tumor. DNA analysis and frozen sections were performed on tumor samples that were taken during surgery. Thirty-one patients met the inclusion criteria for the study. There was a significant difference in G0/G1 phase between high-grade and low-grade tumors. Receiver operating characteristic (ROC) analysis provided 75% of G0/G1 fraction as the optimal cutoff value thresholding the discrimination between low and high-grade tumors. There was a significant difference in S-phase and mitoses fraction between high-grade and low-grade tumors. ROC analysis indicated 6% of S-phase and 9.7% of mitoses as the optimal cutoff values thresholding the discrimination between these two groups. In the glioblastoma patients, we also analyzed the perilesional tissue and found significant differences between tumor mass and margins regarding the G0/G1 phase, the S-phase and mitoses fraction. In conclusion rapid cell cycle analysis is a method capable of differentiating low from high-grade tumors and delineating tumor margins in gliomas. Thus, the role of cell cycle analysis in brain tumors warrants further investigation., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
8. A simple FCM method to avoid misinterpretation in Saccharomyces cerevisiae cell cycle assessment between G0 and sub-G1.
- Author
-
Delobel P and Tesnière C
- Subjects
- DNA metabolism, G1 Phase, Resting Phase, Cell Cycle, Saccharomyces cerevisiae growth & development, Staining and Labeling, Cell Cycle physiology, Flow Cytometry methods, Saccharomyces cerevisiae physiology
- Abstract
Extensively developed for medical and clinical applications, flow cytometry is now being used for diverse applications in food microbiology. Most uses of flow cytometry for yeast cells are derived from methods for mammalian cells, but yeast cells can present specificities that must be taken into account for rigorous analysis of the data output to avoid any misinterpretation. We report an analysis of Saccharomyces cerevisiae cell cycle progression that highlights possible errors. The cell cycle was analyzed using an intercalating fluorochrome to assess cell DNA content. In analyses of yeast cultures, the presence of a sub-G1 peak in the fluorescent signal is often interpreted as a loss of DNA due to its fragmentation associated with apoptosis. However, the cell wall and its stucture may interfere with the fluorescent signal recorded. These observations indicate that misinterpretation of yeast DNA profiles is possible in analyses based on some of the most common probes: cells in G0 appeared to have a lower DNA content and may have been mistaken as a sub-G1 population. However, careful selection of the fluorochrome for DNA quantification allowed a direct discrimination between G0 and G1 yeast cell cycle steps, without additional labeling. We present and discuss results obtained with five current fluorochromes. These observations led us to recommend to use SYTOX Green for cycle analysis of living cells and SYBR Green I for the identification of the apoptosis sub-G1 population identification or the DNA ploidy application.
- Published
- 2014
- Full Text
- View/download PDF
9. Protein tyrosine nitration in the cell cycle.
- Author
-
Jia M, Mateoiu C, and Souchelnytskyi S
- Subjects
- Cell Line, Tumor, G1 Phase, Humans, Proteins chemistry, Resting Phase, Cell Cycle, Tyrosine analysis, Tyrosine metabolism, Cell Cycle, Nitrates metabolism, Protein Processing, Post-Translational, Proteins metabolism, Tyrosine analogs & derivatives
- Abstract
Nitration of tyrosine residues in proteins is associated with cell response to oxidative/nitrosative stress. Tyrosine nitration is relatively low abundant post-translational modification that may affect protein functions. Little is known about the extent of protein tyrosine nitration in cells during progression through the cell cycle. Here we report identification of proteins enriched for tyrosine nitration in cells synchronized in G0/G1, S or G2/M phases of the cell cycle. We identified 27 proteins in cells synchronized in G0/G1 phase, 37 proteins in S phase synchronized cells, and 12 proteins related to G2/M phase. Nineteen of the identified proteins were previously described as regulators of cell proliferation. Thus, our data indicate which tyrosine nitrated proteins may affect regulation of the cell cycle., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
10. CGGBP1 regulates cell cycle in cancer cells.
- Author
-
Singh U, Roswall P, Uhrbom L, and Westermark B
- Subjects
- Animals, Cell Cycle Proteins genetics, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, DNA-Binding Proteins genetics, G1 Phase, GPI-Linked Proteins genetics, Humans, Mice, Neoplasms genetics, Promoter Regions, Genetic, RNA Interference, RNA, Small Interfering genetics, Resting Phase, Cell Cycle, S Phase, Tumor Cells, Cultured, Tumor Suppressor Protein p53 genetics, Cell Cycle, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic, Neoplasms metabolism, Neoplasms pathology
- Abstract
Background: CGGBP1 is a CGG-triplet repeat binding protein, which affects transcription from CGG-triplet-rich promoters such as the FMR1 gene and the ribosomal RNA gene clusters. Earlier, we reported some previously unknown functions of CGGBP1 in gene expression during heat shock stress response. Recently we had found CGGBP1 to be a cell cycle regulatory midbody protein required for normal cytokinetic abscission in normal human fibroblasts, which have all the cell cycle regulatory mechanisms intact., Results: In this study we explored the role of CGGBP1 in the cell cycle in various cancer cell lines. CGGBP1 depletion by RNA interference in tumor-derived cells caused an increase in the cell population at G0/G1 phase and reduced the number of cells in the S phase. CGGBP1 depletion also increased the expression of cell cycle regulatory genes CDKN1A and GAS1, associated with reductions in histone H3 lysine 9 trimethylation in their promoters. By combining RNA interference and genetic mutations, we found that the role of CGGBP1 in cell cycle involves multiple mechanisms, as single deficiencies of CDKN1A, GAS1 as well as TP53, INK4A or ARF failed to rescue the G0/G1 arrest caused by CGGBP1 depletion., Conclusions: Our results show that CGGBP1 expression is important for cell cycle progression through multiple parallel mechanisms including the regulation of CDKN1A and GAS1 levels.
- Published
- 2011
- Full Text
- View/download PDF
11. Viability and cell cycle analysis of equine fibroblasts cultured in vitro.
- Author
-
Lima-Neto JF, Fernandes CB, Alvarenga MA, Golim MA, and Landim-Alvarenga FC
- Subjects
- Animals, Apoptosis, Cells, Cultured, Female, Freezing, G1 Phase, Male, Resting Phase, Cell Cycle, S Phase, Time Factors, Cell Cycle, Cell Survival, Fibroblasts cytology, Horses
- Abstract
This experiment aimed to study equine fibroblasts in culture analyzing and the cell cycle and viability of cells pre- and post-freezing. Skin fragments were obtained from 6 horses and cultured in DMEM high glucose + 10% FCS in 5% CO(2) until the beginning of confluence. Two passages were performed before freezing. Cells subjected to serum starvation (0.5% FCS) were analyzed for viability and cell cycle at 24, 48, 72, 96, 120, 144 and 168 h of culture. For the confluent groups, cells were analyzed at the moment they achieved confluence. Cellular viability was assisted with Hoescht 33342 and propidium iodide. The analysis of apoptosis/necrosis and cell cycle was performed using a flow cytometer (FACS Calibur BD((R))) after staining the cells with annexin V and propidium iodide. Both optical microscopy and flow cytometry confirmed that cellular viability was similar for serum starvation and confluent groups (average 84%). Similarly, both methods were efficient to synchronize the cell cycle before freezing. However, after thawing, serum starvation, for more than 24 h, was superior to culture for synchronizing cells in G0/G1 (69% x 90%). The results of this experiment indicate that equine fibroblasts can be efficiently cultured after thawing.
- Published
- 2010
- Full Text
- View/download PDF
12. Dryopteris crassirhizoma has anti-cancer effects through both extrinsic and intrinsic apoptotic pathways and G0/G1 phase arrest in human prostate cancer cells.
- Author
-
Chang SH, Bae JH, Hong DP, Choi KD, Kim SC, Her E, Kim SH, and Kang CD
- Subjects
- Animals, Antineoplastic Agents, Phytogenic toxicity, BH3 Interacting Domain Death Agonist Protein metabolism, Blotting, Western, Caspases metabolism, Cell Line, Tumor, Dose-Response Relationship, Drug, Drug Synergism, Flow Cytometry, G1 Phase, Gas Chromatography-Mass Spectrometry, Humans, Male, Mice, Mice, Inbred BALB C, Poly(ADP-ribose) Polymerases metabolism, Prostatic Neoplasms metabolism, Resting Phase, Cell Cycle, TNF-Related Apoptosis-Inducing Ligand pharmacology, Time Factors, Antineoplastic Agents, Phytogenic pharmacology, Apoptosis drug effects, Cell Cycle drug effects, Cell Proliferation drug effects, Dryopteris, Prostatic Neoplasms pathology
- Abstract
Aim of the Study: The inhibitory effect of Dryopteris crassirhizoma on the proliferation of human metastatic prostate PC3-MM2 cells and the mechanism of action were examined to identify its anti-cancer properties. The effect of the extract on cell cycle progression and its combined cytotoxic effect with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on PC3-MM2 cells were also investigated., Materials and Methods: The anti-proliferative effects of Dryopteris crassirhizoma were examined by culturing PC3-MM2 cells in the presence or absence of various concentrations of Dryopteris crassirhizoma extract, and the inhibitory effects on cell proliferation were determined by Cell Counting Kit (CCK)-8 analysis. The quantities of apoptosis-inducing proteins were measured by western blotting analysis. Cell cycle progression was analyzed by PI staining using flow cytometry., Results: Dryopteris crassirhizoma (50 and 100 microg/ml) inhibited markedly the proliferation of PC-3 and PC3-MM2 cells without cytotoxicity to normal (spleen) cells from BALB/C mice. Dryopteris crassirhizoma (100 microg/ml) effectively induced apoptosis through the activation of caspase-3, -8, -9, bid, and PARP in PC3-MM2 cells. The cells exposed to Dryopteris crassirhizoma increased significantly the accumulation of the DNA contents in the G0/G1 phase and sub-G1 phase in contrast to the control. The combined cytotoxic effects of Dryopteris crassirhizoma and TRAIL induced the increased activity of 29% in contrast to the sum of the inhibitory effects of each agent alone., Conclusions: Dryopteris crassirhizoma has anti-cancer properties by inducing cell cycle arrest and apoptosis through the extrinsic and intrinsic pathway in PC3-MM2 cells. The extract also showed a combined effect with TRAIL on the inhibition of proliferation in the cells. These findings suggest that possibly its extract could be used for treating androgen-independent prostate cancer with minimal side effects., (Copyright 2010 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
13. Cyclin-C-dependent cell-cycle entry is required for activation of non-homologous end joining DNA repair in postmitotic neurons.
- Author
-
Tomashevski A, Webster DR, Grammas P, Gorospe M, and Kruman II
- Subjects
- Apoptosis, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p21 metabolism, G1 Phase, Hydrogen Peroxide pharmacology, Neurons cytology, Phosphorylation, RNA Interference, RNA, Small Interfering metabolism, Resting Phase, Cell Cycle, Retinoblastoma Protein metabolism, Cell Cycle, Cyclin C metabolism, DNA Repair, Neurons metabolism
- Abstract
It is commonly believed that neurons remain in G(0) phase of the cell cycle indefinitely. Cell-cycle re-entry, however, is known to contribute to neuronal apoptosis. Moreover, recent evidence demonstrates the expression of cell-cycle proteins in differentiated neurons under physiological conditions. The functional roles of such expression remain unclear. Since DNA repair is generally attenuated by differentiation in most cell types, the cell-cycle-associated events in postmitotic cells may reflect the need to re-enter the cell cycle to activate DNA repair. We show that cyclin-C-directed, pRb-dependent G(0) exit activates the non-homologous end joining pathway of DNA repair (NHEJ) in postmitotic neurons. Using RNA interference, we found that abrogation of cyclin-C-mediated exit from G(0) compromised DNA repair but did not initiate apoptosis. Forced G(1) entry combined with prevention of G(1) --> S progression triggered NHEJ activation even in the absence of DNA lesions, but did not induce apoptosis in contrast to unrestricted progression through G(1) --> S. We conclude that G(0) --> G(1) transition is functionally significant for NHEJ repair in postmitotic neurons. These findings reveal the importance of cell-cycle activation for controlling both DNA repair and apoptosis in postmitotic neurons, and underline the particular role of G(1) --> S progression in apoptotic signaling, providing new insights into the mechanisms of DNA damage response (DDR) in postmitotic neurons.
- Published
- 2010
- Full Text
- View/download PDF
14. Transactivators Zta and Rta of Epstein-Barr virus promote G0/G1 to S transition in Raji cells: a novel relationship between lytic virus and cell cycle.
- Author
-
Guo Q, Qian L, Guo L, Shi M, Chen C, Lv X, Yu M, Hu M, Jiang G, and Guo N
- Subjects
- Blotting, Western, Burkitt Lymphoma genetics, Burkitt Lymphoma pathology, Burkitt Lymphoma virology, Butyrates pharmacology, Cell Cycle genetics, Cell Line, Tumor, E2F1 Transcription Factor genetics, Flow Cytometry, G1 Phase, Gene Expression Regulation, Neoplastic drug effects, Gene Expression Regulation, Viral drug effects, Herpesvirus 4, Human genetics, Host-Pathogen Interactions, Humans, Immediate-Early Proteins genetics, Immediate-Early Proteins metabolism, Proteasome Endopeptidase Complex metabolism, Resting Phase, Cell Cycle, Retinoblastoma Protein genetics, Reverse Transcriptase Polymerase Chain Reaction, S Phase, Tetradecanoylphorbol Acetate pharmacology, Trans-Activators genetics, Trans-Activators metabolism, Tumor Suppressor Protein p53 genetics, Virus Activation, Cell Cycle physiology, Herpesvirus 4, Human physiology, Immediate-Early Proteins physiology, Trans-Activators physiology
- Abstract
In the present study, we show that the treatment of Epstein-Barr virus (EBV) latently infected Raji cells with TPA/SB caused the cell growth arrest. The Zta-positive cells were predominantly enriched in G0/G1 phase of cell cycle. When Zta expression reached a maximal level, a fraction of Zta expressing cell population reentered S phase. Analysis of the expression pattern of a key set of cell cycle regulators revealed that the expression of Zta and Rta substantially interfered with the cell cycle regulatory machinery in Raji cells, strongly inhibiting the expression of Rb and p53 and inducing the expression of E2F1. Down-regulation of Rb was further demonstrated to be mediated by proteasomal degradation, and p53 and p21 affected at transcription level. The data indicate that both Zta and Rta promote entry into S phase of Raji cells. The important roles of Zta and Rta in EBV lytic reactivation were also demonstrated. Our finding suggests that these two transcriptional activators may act synergistically to govern the expression of downstream early and late genes as well as cellular genes and initiation of lytic cycle and manipulation of cell cycle regulatory mechanisms require the joint and interactive contributions of Rta and Zta., ((c) 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
15. MFSD2A is a novel lung tumor suppressor gene modulating cell cycle and matrix attachment.
- Author
-
Spinola M, Falvella FS, Colombo F, Sullivan JP, Shames DS, Girard L, Spessotto P, Minna JD, and Dragani TA
- Subjects
- Animals, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Cell Adhesion genetics, Cell Line, Tumor, Cell Movement genetics, Cell Proliferation, Down-Regulation genetics, Extracellular Matrix genetics, Female, G1 Phase, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Immunohistochemistry, Lung Neoplasms genetics, Lung Neoplasms pathology, Membrane Transport Proteins metabolism, Mice, Mice, Nude, Resting Phase, Cell Cycle, Symporters, Tumor Suppressor Proteins metabolism, Cell Cycle genetics, Extracellular Matrix metabolism, Genes, Tumor Suppressor, Membrane Transport Proteins genetics, Tumor Suppressor Proteins genetics
- Abstract
Background: MFSD2A (major facilitator superfamily domain containing 2) gene maps on chromosome 1p34 within a linkage disequilibrium block containing genetic elements associated with progression of lung cancer., Results: Here we show that MFSD2A expression is strongly downregulated in non-small cell lung cancer cell lines of different histotypes and in primary lung adenocarcinomas. Experimental modulation of MFSD2A in lung cancer cells is associated with alteration of mRNA levels of genes involved in cell cycle control and interaction with the extracellular matrix. Exogenous expression of MFSD2A in lung cancer cells induced a G1 block, impaired adhesion and migration in vitro, and significantly reduced tumor colony number in vitro (4- to 27-fold, P < 0.0001) and tumor volume in vivo (approximately 3-fold, P < 0.0001). siRNA knockdown studies in normal human bronchial epithelial cells confirmed the role of MFSD2A in G1 regulation., Conclusion: Together these data suggest that MFSD2A is a novel lung cancer tumor suppressor gene that regulates cell cycle progression and matrix attachment.
- Published
- 2010
- Full Text
- View/download PDF
16. Cell cycle-dependent regulation of the bi-directional overlapping promoter of human BRCA2/ZAR2 genes in breast cancer cells.
- Author
-
Misra S, Sharma S, Agarwal A, Khedkar SV, Tripathi MK, Mittal MK, and Chaudhuri G
- Subjects
- 5' Untranslated Regions genetics, Animals, BRCA2 Protein metabolism, Cell Line, Tumor, Cell Nucleus metabolism, Codon genetics, Conserved Sequence, Cytosol metabolism, Exons genetics, Female, G1 Phase, Gene Knockdown Techniques, Gene Rearrangement genetics, Humans, Introns genetics, Protein Transport, RNA, Messenger genetics, RNA, Messenger metabolism, Resting Phase, Cell Cycle, Sequence Homology, Amino Acid, Transcription Factors metabolism, Vertebrates genetics, BRCA2 Protein genetics, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Cycle genetics, Gene Expression Regulation, Neoplastic, Promoter Regions, Genetic, Transcription Factors genetics
- Abstract
Background: BRCA2 gene expression is tightly regulated during the cell cycle in human breast cells. The expression of BRCA2 gene is silenced at the G0/G1 phase of cell growth and is de-silenced at the S/G2 phase. While studying the activity of BRCA2 gene promoter in breast cancer cells, we discovered that this promoter has bi-directional activity and the product of the reverse activity (a ZAR1-like protein, we named ZAR2) silences the forward promoter at the G0/G1 phase of the cell. Standard techniques like cell synchronization by serum starvation, flow cytometry, N-terminal or C-terminal FLAG epitope-tagged protein expression, immunofluorescence confocal microscopy, dual luciferase assay for promoter evaluation, and chromatin immunoprecipitation assay were employed during this study., Results: Human BRCA2 gene promoter is active in both the forward and the reverse orientations. This promoter is 8-20 fold more active in the reverse orientation than in the forward orientation when the cells are in the non-dividing stage (G0/G1). When the cells are in the dividing state (S/G2), the forward activity of the promoter is 5-8 folds higher than the reverse activity. The reverse activity transcribes the ZAR2 mRNA with 966 nt coding sequence which codes for a 321 amino acid protein. ZAR2 has two C4 type zinc fingers at the carboxyl terminus. In the G0/G1 growth phase ZAR2 is predominantly located inside the nucleus of the breast cells, binds to the BRCA2 promoter and inhibits the expression of BRCA2. In the dividing cells, ZAR2 is trapped in the cytoplasm., Conclusions: BRCA2 gene promoter has bi-directional activity, expressing BRCA2 and a novel C4-type zinc finger containing transcription factor ZAR2. Subcellular location of ZAR2 and its expression from the reverse promoter of the BRCA2 gene are stringently regulated in a cell cycle dependent manner. ZAR2 binds to BRCA2/ZAR2 bi-directional promoter in vivo and is responsible, at least in part, for the silencing of BRCA2 gene expression in the G0/G1 phase in human breast cells.
- Published
- 2010
- Full Text
- View/download PDF
17. Serum starvation and full confluency for cell cycle synchronization of domestic cat (felis catus) foetal fibroblasts.
- Author
-
de Barros FR, Goissis MD, Caetano HV, Paula-Lopes FF, Peres MA, Assumpção ME, and Visintin JA
- Subjects
- Animals, Cloning, Organism veterinary, DNA analysis, Fibroblasts chemistry, Flow Cytometry veterinary, G1 Phase, Nuclear Transfer Techniques veterinary, Resting Phase, Cell Cycle, Cats embryology, Cell Cycle physiology, Culture Media, Serum-Free, Fibroblasts ultrastructure
- Abstract
Nuclear transfer of domestic cat can be used as a tool to develop reproductive biotechnologies in wild felids. The importance of cell cycle phase during the nuclear transfer has been a matter of debate since the first mammalian clone was produced. The cell cycle phase of donor cells interferes on maintenance of correct ploidy and genetic reprogramming of the reconstructed embryo. The use of G0/G1 arrested donor cells has been shown to improve nuclear transfer efficiency. The present study was conducted to test the hypothesis that domestic cat foetal fibroblasts cultured up to the fifth passage and submitted to full confluency provide a higher percentage of cells at G0/G1 stage than fibroblasts cultured in serum starved media. Results demonstrated that serum starvation increased (p < or = 0.05) the percentage of G0/G1 fibroblasts when compared with control. Moreover, the combined protocol using confluency and serum starvation was more efficient (p < or = 0.05) synchronizing cells at G0/G1 stage than serum starvation or confluency alone for the first 3 days of treatment. In conclusion, serum starvation and full confluency act in a synergistic manner to improve domestic cat foetal fibroblast cell cycle synchronization at the G0/G1 stage.
- Published
- 2010
- Full Text
- View/download PDF
18. Anti-proliferative effects of gamma-tocotrienol on mammary tumour cells are associated with suppression of cell cycle progression.
- Author
-
Samant GV, Wali VB, and Sylvester PW
- Subjects
- Animals, Cell Line, Cyclin D1 metabolism, Cyclin-Dependent Kinase 2 metabolism, Cyclin-Dependent Kinase 4 metabolism, Cyclin-Dependent Kinase 6 metabolism, Cyclin-Dependent Kinase Inhibitor p27 metabolism, Epidermal Growth Factor pharmacology, Female, Mammary Glands, Animal metabolism, Mice, Phosphorylation, Resting Phase, Cell Cycle, Vitamin E pharmacology, Cell Cycle drug effects, Chromans pharmacology, Mammary Neoplasms, Experimental metabolism, Vitamin E analogs & derivatives
- Abstract
Objectives: Previous studies have shown that gamma-tocotrienol induces potent anti-proliferative effects on +SA mammary tumour cells in culture; here, investigations have been conducted to determine its effects on intracellular signalling proteins involved in regulating cell cycle progression., Materials and Methods: +SA cells were maintained in mitogen-free defined media containing 0 or 4 micromgamma-tocotrienol, for 48 h to synchronize cell cycle in G(0) phase, and then they were exposed to 100 ng/ml EGF to initiate cell cycle progression. Whole cell lysates were collected at various time points from each treatment group and were prepared for Western blot analysis., Results and Conclusions: Treatment with 4 micromgamma-tocotrienol significantly inhibited +SA cell proliferation over a 4-day culture period. Moreover, this treatment resulted in a relatively large reduction in cyclin D1, cyclin dependent kinase (CDK)4, CDK2 and CDK6 levels, between 4 and 24 h after EGF exposure. Tocotrienol treatment also resulted in a relatively large increase in CDK inhibitor (CKI) p27, prior to and after EGF exposure, but had little effect on levels of CKIs, p21 and p15. Tocotrienol treatment also induced a large relative reduction in retinoblastoma (Rb) protein phosphorylation at ser780 and ser807/811. These findings strongly suggest that anti-proliferative effects of gamma-tocotrienol are associated with reduction in cell cycle progression from G(1) to S, as evidenced by increased p27 levels, and a corresponding decrease in cyclin D1, CDK2, CDK4, CDK6 and phosphorylated Rb levels.
- Published
- 2010
- Full Text
- View/download PDF
19. Cloning and characterization of a novel intracellular protein p48.2 that negatively regulates cell cycle progression.
- Author
-
Yang F, Xu YP, Li J, Duan SS, Fu YJ, Zhang Y, Zhao Y, Qiao WT, Chen QM, Geng YQ, Che CY, Cao YL, Wang Y, Zhang L, Long L, He J, Cui QC, Chen SC, Wang SH, and Liu L
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Cell Line, Cloning, Molecular, Computational Biology, Cyclin D1 genetics, Cyclin D1 metabolism, Cyclin D3 genetics, Cyclin D3 metabolism, Down-Regulation genetics, G1 Phase, Gene Expression Profiling, Gene Expression Regulation, Genome, Human genetics, Humans, Mice, Molecular Sequence Data, Promoter Regions, Genetic genetics, RNA, Small Interfering, Receptors, Cytokine chemistry, Receptors, Cytokine metabolism, Resting Phase, Cell Cycle, STAT3 Transcription Factor metabolism, Sequence Homology, Amino Acid, Signal Transduction, Cell Cycle genetics, Cell Cycle Proteins genetics, Intracellular Space metabolism, Receptors, Cytokine genetics
- Abstract
Neurofibromatosis type 1 (NF1) microdeletion is a large genomic deletion that embraces at least 11 continuous genes at human chromosome 17q11.2. To date, most of these genes' functions still remain undefined. In this study, we report an unknown cytokine receptor like molecule (p48.2) that is frequently deleted in patients with type-1 and type-2 NF1 microdeletions in the neurofibromin locus. The cloned gene has 1317 base pair long that encodes a 438aa intracellular protein. The gene was subsequently named p48.2 based on its predicted molecular weight. A typical fibronectin type III (FNIII) domain was identified in p48.2 between Arg(176) and Pro(261) in which a palindromic Arg-Gly-Asp (RGD) repeat plus a putative Trp-Ser-X-Trp-Ser (WSXWS) motif were found at the domain's C-terminus. p48.2 mRNAs were abundant in many tumor cell lines and normal human tissues and up-regulated in some freshly isolated lung cancer and leukemia cells. Interestingly, over-expression of p48.2 in human embryo kidney 293T cells could significantly cause G0/G1 arrest and prevented S phase entry. In contrast, repressing endogenous p48.2 gene expression by specific siRNA markedly reduced G0/G1 population. Importantly, over-expression of p48.2 could significantly up-regulate rather than down-regulate cyclin D1 and cyclin D3 expressions. We further showed that the induction of cyclin D1 expression was directly due to the activation of signal transducers and activators of transcription 3 (STAT3), but was independent of RAS/mitogen-activated protein kinase (RAS/MAPK) signaling pathway. Thus, p48.2 may represent a novel type of intracellular protein functioning as a negative regulator at the G0/G1 phase.
- Published
- 2009
- Full Text
- View/download PDF
20. High GATA-2 expression inhibits human hematopoietic stem and progenitor cell function by effects on cell cycle.
- Author
-
Tipping AJ, Pina C, Castor A, Hong D, Rodrigues NP, Lazzari L, May GE, Jacobsen SE, and Enver T
- Subjects
- Animals, Apoptosis, Cells, Cultured cytology, Cells, Cultured drug effects, Cord Blood Stem Cell Transplantation, Estradiol pharmacology, Fetal Blood cytology, GATA2 Transcription Factor biosynthesis, GATA2 Transcription Factor genetics, Gene Expression Regulation genetics, Genes, Synthetic, Genes, cdc, Humans, Interleukin-3 pharmacology, Mice, Mice, Inbred NOD, Mice, SCID, Receptors, Estrogen drug effects, Receptors, Estrogen genetics, Recombinant Fusion Proteins physiology, Resting Phase, Cell Cycle, Tamoxifen pharmacology, Transcription, Genetic, Cell Cycle, GATA2 Transcription Factor physiology, Hematopoietic Stem Cells cytology
- Abstract
Evidence suggests the transcription factor GATA-2 is a critical regulator of murine hematopoietic stem cells. Here, we explore the relation between GATA-2 and cell proliferation and show that inducing GATA-2 increases quiescence (G(0) residency) of murine and human hematopoietic cells. In human cord blood, quiescent fractions (CD34(+)CD38(-)Hoechst(lo)Pyronin Y(lo)) express more GATA-2 than cycling counterparts. Enforcing GATA-2 expression increased quiescence of cord blood cells, reducing proliferation and performance in long-term culture-initiating cell and colony-forming cell (CFC) assays. Gene expression analysis places GATA-2 upstream of the quiescence regulator MEF, but enforcing MEF expression does not prevent GATA-2-conferred quiescence, suggesting additional regulators are involved. Although known quiescence regulators p21(CIP1) and p27(KIP1) do not appear to be responsible, enforcing GATA-2 reduced expression of regulators of cell cycle such as CCND3, CDK4, and CDK6. Enforcing GATA-2 inhibited human hematopoiesis in vivo: cells with highest exogenous expression (GATA-2(hi)) failed to contribute to hematopoiesis in nonobese diabetic-severe combined immunodeficient (NOD-SCID) mice, whereas GATA-2(lo) cells contributed with delayed kinetics and low efficiency, with reduced expression of Ki-67. Thus, GATA-2 activity inhibits cell cycle in vitro and in vivo, highlighting GATA-2 as a molecular entry point into the transcriptional program regulating quiescence in human hematopoietic stem and progenitor cells.
- Published
- 2009
- Full Text
- View/download PDF
21. Different culture conditions used for arresting the G0/G1 phase of the cell cycle in goldfish (Carassius auratus) caudal fin-derived fibroblasts.
- Author
-
Choresca CH Jr, Koo OJ, Oh HJ, Hong SG, Gomez DK, Kim JH, Lee BC, and Park SC
- Subjects
- Analysis of Variance, Animals, Cell Culture Techniques, Cell Division drug effects, Cells, Cultured, Fibroblasts drug effects, G1 Phase, Goldfish, Purines metabolism, Purines pharmacology, Resting Phase, Cell Cycle, Roscovitine, Starvation, Cell Cycle, Fibroblasts cytology
- Abstract
One of the most important factors determining the success of the development of cloned embryos is the cell cycle stage of the donor cells. We investigated the effects of serum starvation, culturing to confluence and roscovitine treatment on the cell cycle synchronization of goldfish caudal fin-derived fibroblasts by flow cytometric analysis. The results show that culturing the cells to confluence (85.5%) and roscovitine treatment (82.71%) yield a significantly higher percentage of cells arrested in the G0/G1 (P<0.05) phase than serum starvation (62.85%). Different concentrations of roscovitine (5, 10, or 15 microM) induce cell cycle arrest at the G0/G1 phase.
- Published
- 2009
- Full Text
- View/download PDF
22. The potential role of ribosomal protein S5 on cell cycle arrest and initiation of murine erythroleukemia cell differentiation.
- Author
-
Matragkou CN, Papachristou ET, Tezias SS, Tsiftsoglou AS, Choli-Papadopoulou T, and Vizirianakis IS
- Subjects
- Animals, Apoptosis, Cyclin-Dependent Kinases analysis, G1 Phase, Mice, Resting Phase, Cell Cycle, Ribosomal Proteins genetics, Transfection, Cell Cycle, Cell Differentiation, Leukemia, Erythroblastic, Acute pathology, Ribosomal Proteins pharmacology
- Abstract
Evidence now exists to indicate that some ribosomal proteins besides being structural components of the ribosomal subunits are involved in the regulation of cell differentiation and apoptosis. As we have shown earlier, initiation of erythroid differentiation of murine erythroleukemia (MEL) cells is associated with transcriptional inactivation of genes encoding ribosomal RNAs and ribosomal proteins S5 (RPS5) and L35a. In this study, we extended these observations and investigated whether transfection of MEL cells with RPS5 cDNA affects the onset of initiation of erythroid maturation and their entrance in cell cycle arrest. Stably transfected MEL cloned cells (MEL-C14 and MEL-C56) were established and assessed for their capacity to produce RPS5 RNA transcript and its translated product. The impact of RPS5 cDNA transfection on the RPS5 gene expression patterns and the accumulation of RPS5 protein in inducible transfected MEL cells were correlated with their ability to: (a) initiate differentiation, (b) enter cell cycle arrest at G(1)/G(0) phase, and (c) modulate the level of cyclin-dependent kinases CDK2, CDK4, and CDK6. The data presented indicate that deregulation of RPS5 gene expression (constitutive expression) affects RPS5 protein level and delays both the onset of initiation of erythroid maturation and entrance in cell cycle arrest in inducer-treated MEL cells., (2008 Wiley-Liss, Inc.)
- Published
- 2008
- Full Text
- View/download PDF
23. Prohibitin silencing reverses stabilization of mitochondrial integrity and chemoresistance in ovarian cancer cells by increasing their sensitivity to apoptosis.
- Author
-
Gregory-Bass RC, Olatinwo M, Xu W, Matthews R, Stiles JK, Thomas K, Liu D, Tsang B, and Thompson WE
- Subjects
- Adenoviridae, Blotting, Western, Carcinoma, Endometrioid drug therapy, Carcinoma, Papillary drug therapy, Caspase 3 metabolism, Cell Cycle drug effects, Cell Line, Tumor, Cell Survival drug effects, Cell Survival genetics, Drug Resistance, Neoplasm, Enzyme Inhibitors pharmacology, Female, G1 Phase, Gene Expression Regulation, Neoplastic drug effects, Genetic Vectors, Humans, Ki-67 Antigen analysis, Microscopy, Fluorescence, Ovarian Neoplasms drug therapy, Ovary chemistry, Prohibitins, RNA, Small Interfering metabolism, Recombinant Proteins, Repressor Proteins metabolism, Resting Phase, Cell Cycle, Staurosporine pharmacology, Up-Regulation, Antineoplastic Agents pharmacology, Apoptosis drug effects, Apoptosis genetics, Carcinoma, Endometrioid genetics, Carcinoma, Papillary genetics, Cell Cycle genetics, Gene Silencing, Mitochondria metabolism, Ovarian Neoplasms genetics, Repressor Proteins genetics
- Abstract
Current approaches to the treatment of ovarian cancer are limited because of the development of resistance to chemotherapy. Prohibitin (Phb1) is a possible candidate protein that contributes to development of drug resistance, which could be targeted in neoplastic cells. Phb1 is a highly conserved protein that is associated with a block in the G0/G1 phase of the cell cycle and also with cell survival. Our study was designed to determine the role of Phb1 in regulating cellular growth and apoptosis in ovarian cancer cells. Our results showed that Phb1 content is differentially overexpressed in papillary serous ovarian carcinoma and endometrioid ovarian adenocarcinoma when compared to normal ovarian epithelium and was inversely related to Ki67 expression. Immunofluorescence microscopy and Western analyses revealed that Phb1 is primarily associated with the mitochondria in ovarian cancer cells. Over-expression of Phb1 by adenoviral Phb1 infection resulted in an increase in the percentage of ovarian cancer cells accumulating at G0/G1 phase of the cell cycle. Treatment of ovarian cancer cells with staurosporine (STS) induced apoptosis in a time-dependent manner. Phb1 over-expression induced cellular resistance to STS via the intrinsic apoptotic pathway. In contrast, silencing of Phb1 expression by adenoviral small interfering RNA (siRNA) sensitized ovarian cancer cells to STS-induce apoptosis. Taken together, these results suggest that Phb1 induces block at G0/G1 phase of the cell cycle and promotes survival of cancer cells. Furthermore, silencing of the Phb1 gene expression may prove to be a valuable therapeutic approach for chemoresistant ovarian cancer by increasing sensitivity of cancer cells to apoptosis., ((c) 2008 Wiley-Liss, Inc.)
- Published
- 2008
- Full Text
- View/download PDF
24. Effects of serum deprivation and cycloheximide on cell cycle of low and high passage porcine fetal fibroblasts.
- Author
-
Goissis MD, Caetano HV, Marques MG, de Barros FR, Feitosa WB, Milazzotto MP, Binelli M, Assumpção ME, and Visintin JA
- Subjects
- Animals, Cell Cycle physiology, Cell Division physiology, Cell Separation veterinary, Cell Survival, Cells, Cultured, Fibroblasts cytology, Fibroblasts drug effects, Flow Cytometry veterinary, G1 Phase, Resting Phase, Cell Cycle, Cell Cycle drug effects, Culture Media, Serum-Free pharmacology, Cycloheximide pharmacology, Fibroblasts physiology, Swine embryology
- Abstract
Arrest of cells in G0/G1 cell cycle phase is desired for nuclear transfer procedures. Serum starvation and cell cycle inhibitors are different ways to induce synchronization of the cell cycle. This study investigated the effects of serum starvation and cycloheximide (CHX) on the cell cycle of low (5th) and high (15th) passages fetal porcine fibroblasts. Cell cycle phases were determined using fluorescent activated cell sorting. Fifth passage fibroblast cultures had higher (p < 0.05) proportion of cells in G0/G1 only after 72 h of serum starvation (77.60 +/- 0.65) when compared with non-starved cells (71.44 +/- 1.88). Serum starvation for all periods tested induced an increase (p < 0.05) on proportion of cells in G0/G1 on the 15th passage. No significant differences were observed on the 5th passage cultures exposed to CHX, although, on the 15th passage an increase on proportion of cells was observed after all periods of exposure (p < 0.05). These data indicates that high passage cells in vitro are more susceptible to serum starvation and CHX G0/G1 synchronization.
- Published
- 2007
- Full Text
- View/download PDF
25. The cyclin-dependent kinase inhibitors p15INK4B and p21CIP1 are critical regulators of fibrillar collagen-induced tumor cell cycle arrest.
- Author
-
Wall SJ, Zhong ZD, and DeClerck YA
- Subjects
- Cyclin-Dependent Kinase Inhibitor p27, G2 Phase, Humans, Resting Phase, Cell Cycle, Cell Cycle, Cyclin-Dependent Kinase Inhibitor p15 physiology, Cyclin-Dependent Kinase Inhibitor p21 physiology, Fibrillar Collagens physiology, Melanoma pathology
- Abstract
The extracellular matrix is a crucial component in determining cell fate. Fibrillar collagen in its native form inhibits cell proliferation, whereas in its monomeric form it stimulates proliferation. The observation of elevated levels of p27(KIP1) in cells plated in the presence of fibrillar collagen has led to the assumption that this kinase inhibitor was responsible for cell cycle arrest on fibrillar collagen. Here we provide evidence that p15(INK4b), rather than p27(KIP1), is the cyclin-dependent kinase inhibitor responsible for G0/G1 arrest of human melanoma cells grown on fibrillar collagen. Additionally, we demonstrate that fibrillar collagen can also arrest cells at the G2 phase, which is mediated in part by p21(CIP1). Our data, in addition to identifying cyclin-dependent kinase inhibitors important in cell cycle arrest mediated by fibrillar collagen, demonstrate the complexity of cell cycle regulation and indicate that modulating a single cyclin-dependent kinase inhibitor does not disrupt cell proliferation in the presence of fibrillar collagen.
- Published
- 2007
- Full Text
- View/download PDF
26. [Effects of serum starvation and contact inhibition on the cell cycle G0 synchronization of the human embryonic lung fibroblast].
- Author
-
Yan L and Tao M
- Subjects
- Cells, Cultured, Embryo, Mammalian, Flow Cytometry, G2 Phase, Humans, Resting Phase, Cell Cycle, Cell Culture Techniques methods, Cell Cycle physiology, Culture Media, Serum-Free pharmacology, Fibroblasts cytology, Lung cytology
- Abstract
Objective: To compare the effects of two different cell cycle G0 stage synchronization methods in the human embryonic lung fibroblast and re-entry into the cell cycle., Methods: Cell cycle synchronization was achieved by serum starvation and contact inhibition. Which were identified by flow cytometry. Cell morphologic character were also analyzed. Detection cell cycle distribution change after serum starvation(0.5% serum) for 48h then serum re-stimulation 24h, growth in contact inhibition 72h and subsequent passage 0-24h respectively., Results: Serum starvation 48h could well synchronize HELF in G0 stage. Re-stimulated by serum 16h cell re-entered cycle and S increased to peak at 20h, G2 increased to peak at 24h. Growth in contact inhibition 72h could synchronize 83.36% cell in G0. Cells re-entered cycle after passage 13h and S increased to peak at 22h, G2 increased to peak at 25h., Conclusion: Serum starvation 48h and growth in confluence 72h both can synchronize HELF in G0 stage effectively. Cells could re-enter cell cycle 16h after re-stimulated by serum or 13h after passage respectively. Serum starvation method was better than contact inhibition.
- Published
- 2007
27. Beta-hydroxyisovalerylshikonin induces apoptosis and G0/G1 cell-cycle arrest of endometriotic stromal cells: a preliminary in vitro study.
- Author
-
Nishida M, Nasu K, Ueda T, Yuge A, Takai N, and Narahara H
- Subjects
- Bromodeoxyuridine, Cell Division drug effects, Cell Survival drug effects, DNA Fragmentation, Female, G1 Phase, G2 Phase, Humans, In Vitro Techniques, Premenopause, Resting Phase, Cell Cycle, S Phase, Stromal Cells drug effects, Apoptosis drug effects, Cell Cycle drug effects, Endometriosis pathology, Naphthoquinones pharmacology, Stromal Cells pathology
- Abstract
Background: Most of the current medical treatments for endometriosis aim to down-regulate the estrogen activity. However, a high recurrence rate after medical treatments has been the most significant problem. Beta-hydroxyisovalerylshikonin (beta-HIVS) is an ATP non-competitive inhibitor of protein-tyrosine kinases and is considered an apoptosis-inducing agent. The aim of this study is to evaluate the effects of beta-HIVS on the proliferation, cell cycle and apoptosis of endometriotic stromal cells., Methods: We investigated the effects of beta-HIVS on cultured ovarian endometriotic cyst stromal cells (ECSC) by a modified methylthiazoletetrazolium (MTT) assay, a 5-bromo-2'-deoxyuridine (BrdU) incorporation assay and internucleosomal DNA fragmentation assays. The effect of beta-HIVS on the cell cycle of ECSC was determined by flow cytometry. The expression of apoptosis-related molecules was examined in ECSC using western blot analysis., Results: Beta-HIVS significantly inhibited the proliferation and DNA synthesis of ECSC and induced apoptosis and G0/G1 phase cell-cycle arrest of these cells. Down-regulation of the B-cell lymphoma/leukaemia-2 (Bcl-2) expression with the activation of caspase-3, caspase-8 and caspase-9 was observed in ECSC after beta-HIVS treatment., Conclusions: These results suggest that beta-HIVS induces apoptosis of ECSC by suppressing anti-apoptotic proteins. Although our present findings are preliminary, beta-HIVS could potentially be a therapeutic agent for the treatment of endometriosis.
- Published
- 2006
- Full Text
- View/download PDF
28. MDDD, a 4,9-diazapyrenium derivative, is selectively toxic to glioma cells by inducing growth arrest at G0/G1 independently of p53.
- Author
-
Yu DH, Macdonald J, Josephs S, Liu Q, Nguy V, Tor Y, Wong-Staal F, and Li QX
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, DNA drug effects, DNA metabolism, DNA Damage, G1 Phase, Glioma genetics, Glioma metabolism, Humans, Inhibitory Concentration 50, Neoplastic Stem Cells drug effects, RNA Interference, Rats, Resting Phase, Cell Cycle, Tumor Stem Cell Assay, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Antineoplastic Agents pharmacology, Cell Cycle drug effects, Glioma pathology, Intercalating Agents pharmacology, Pyridinium Compounds pharmacology
- Abstract
4-Methyl-2,7-diamino-5,10-diphenyl-4,9-diaz-apyrenium chloride (MDDD), a stable and water soluble nucleic acid-intercalating agent, was shown to be toxic to cancer cells with IC50 around 10 microM. IC(50) We tested MDDD for its potential antitumor activities and found it inhibited cancer cell growth with IC(50) in the micromolar range for the majority of cancer cells tested, with the exception of glioma cells, for which the IC(50) is in the submicromolar range. This unique selectivity of MDDD to glioma cells can potentially be exploited for anti-glioma therapeutics. Although the underlying mechanisms for the apparent glioma specificity remain to be elucidated, our analysis indicates that MDDD significantly reduces cell clonogenicity and blockes cell proliferation at the G1 phase. MDDD treatment also triggers induction of p53 and p21 at the protein levels, suggesting the activation of DNA damage response. However, MDDD mediated growth inhibition does not require the p53 pathway since p53+/- isogenic cell pairs display the same sensitivity. These properties of MDDD favor its candidacy for evaluation as a new anti-tumor agent, particularly for glioma.
- Published
- 2006
- Full Text
- View/download PDF
29. [Mechanism of cell proliferation--cell cycle, oncogenes, and senescence].
- Author
-
Ide T
- Subjects
- Animals, Antineoplastic Agents, Cell Survival, Cyclin-Dependent Kinase Inhibitor p21 physiology, Drug Design, Histone Acetyltransferases physiology, Humans, Mitogen-Activated Protein Kinases physiology, Neoplasms drug therapy, Neoplasms genetics, Resting Phase, Cell Cycle, Signal Transduction physiology, Tumor Suppressor Protein p53 physiology, Cell Cycle, Cell Proliferation, Cellular Senescence, Oncogenes, Telomerase physiology, Telomere pathology
- Abstract
Cell proliferation is regulated through a transition between the G0 phase and cell cycle. We isolated a mammalian temperature-sensitive mutant cell line defective in the function from the G0 phase to cell cycle. Senescent human somatic cells fail to enter into the cell cycle from the G0 phase with stimulation by any growth factor. Telomere shortening was found to be a cause of cellular senescence, and reexpression of telomerase immortalized human somatic cells. Immortalized human somatic cells showed normal phenotypes and were useful not only for basic research but also for clinical and applied fields. The importance of p53 and p21 activation/induction i now well accepted in the signal transduction process from telomere shortening to growth arrest, but the precise mechanism is largely unknown as yet. We found that the MAP kinase cascade and histone acetylase have an important role in the signaling process to express p21. Tumor tissues and cells were found to have strong telomerase activity, while most normal somatic human tissues showed very weak or no activity. Telomerase activity was shown to be a good marker for early tumor diagnosis because significant telomerase activity was detected in very early tumors or even in some precancerous tissues compared with adjacent normal tissues. Telomere/telomerase is a candidate target for cancer chemotherapeutics, and an agent that abrogated telomere functions was found to kill tumor cells effectively by inducing apoptosis whereas it showed no effect on the viability of normal cells.
- Published
- 2006
- Full Text
- View/download PDF
30. Labd-14-ene-8,13-diol (sclareol) induces cell cycle arrest and apoptosis in human breast cancer cells and enhances the activity of anticancer drugs.
- Author
-
Dimas K, Papadaki M, Tsimplouli C, Hatziantoniou S, Alevizopoulos K, Pantazis P, and Demetzos C
- Subjects
- Breast Neoplasms pathology, Cell Line, Tumor, Cisplatin pharmacology, Dose-Response Relationship, Drug, Doxorubicin pharmacology, Drug Interactions, Etoposide pharmacology, Female, G1 Phase, Humans, Resting Phase, Cell Cycle, S Phase, Time Factors, Tumor Suppressor Protein p53 metabolism, Antineoplastic Agents pharmacology, Apoptosis drug effects, Breast Neoplasms metabolism, Cell Cycle, Cell Proliferation drug effects, Diterpenes pharmacology
- Abstract
Sclareol is a labdane-type diterpene that has demonstrated a significant cytotoxic activity against human leukemic cell lines. Here, we report the effect of sclareol against the human breast cancer cell lines MN1 and MDD2 derived from the parental cell line, MCF7. MN1 cells express functional p53, whereas MDD2 cells do not express p53. Flow cytometry analysis of the cell cycle indicated that sclareol was able to inhibit DNA synthesis induce arrest at the G(0/1) phase of the cycle apoptosis independent of p53. Sclareol-induced apoptosis was further assessed by detection of fragmented DNA in the cells. Furthermore, sclareol enhanced the activity of known anticancer drugs, doxorubicin, etoposide and cisplatinum, against MDD2 breast cancer cell line.
- Published
- 2006
- Full Text
- View/download PDF
31. Heme oxygenase overexpression attenuates glucose-mediated oxidative stress in quiescent cell phase: linking heme to hyperglycemia complications.
- Author
-
Sacerdoti D, Olszanecki R, Li Volti G, Colombrita C, Scapagnini G, and Abraham NG
- Subjects
- Cells, Cultured, Computer Systems, Culture Media, Serum-Free, Dinoprost analogs & derivatives, Dinoprost biosynthesis, G1 Phase, Heme metabolism, Heme Oxygenase (Decyclizing) genetics, Humans, Hyperglycemia blood, Hyperglycemia complications, Oxidative Stress physiology, RNA, Messenger metabolism, Resting Phase, Cell Cycle, Reverse Transcriptase Polymerase Chain Reaction, Superoxides metabolism, Cell Cycle physiology, Endothelial Cells cytology, Endothelial Cells metabolism, Glucose pharmacology, Heme Oxygenase (Decyclizing) metabolism, Oxidative Stress drug effects
- Abstract
Heme oxygenase (HO-1) is a stress protein, which has been suggested to participate in defense mechanisms against glucose induced oxidative injury. The purpose of this study was to examine the role of human HO-1 in attenuating glucose-mediated oxidative stress. We investigated the effect of high ambient glucose (15, 33 and 66 mM) on HO-1 gene expression in endothelial cells grown in a serum deprived media compared to the effect of glucose on exponentially grown cells (10% FBS). High glucose at 15 and 33 mM caused significant inhibition of HO-1 protein and activity in G0/G1 and in cells exponentially grown. Glucose concentration at 66 mM caused a significant increase in HO-1. Addition of heme (10 microM) increased HO-1 protein and bilirubin formation in G0/G1, in a time dependent manner peaking at 16 h. Glucose attenuated heme mediated increase in HO-1 proteins. RT-PCR demonstrated that glucose decreased the levels of HO-1 mRNA in both G0/G1 or cells grown in 10% FBS. The rate of HO-1 induction in response to heme was several fold higher in serum-starved cells compared to cells cultured in 10% FBS. Cells exposed to high glucose for up to 24 h had a significant increase in cellular heme and potentiated heme-mediated increase in generation of superoxide anion and 8-epi-isoprostane PGF(2alpha). HO-1 gene transduction prevented glucose-mediated elevation of 8-epi-isoprostane PGF(2alpha). These results imply that expression of HO-1 in G0/G1 cells may be a key player in decreasing cellular heme, associated with increased generation of bilirubin, and in attenuating glucose mediated oxidative stress.
- Published
- 2005
- Full Text
- View/download PDF
32. The antiproliferative activity of prodelphinidin B-2 3'-O-gallate from green tea leaf is through cell cycle arrest and Fas-mediated apoptotic pathway in A549 cells.
- Author
-
Kuo PL, Hsu YL, Lin TC, and Lin CC
- Subjects
- Antineoplastic Agents, Phytogenic therapeutic use, Cell Division drug effects, Enzyme-Linked Immunosorbent Assay, Fas Ligand Protein, Flow Cytometry, G1 Phase, Humans, Lung Neoplasms drug therapy, Membrane Glycoproteins, Resting Phase, Cell Cycle, Tumor Cells, Cultured, Anthocyanins pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Apoptosis drug effects, Cell Cycle drug effects, Tea chemistry, Tumor Suppressor Protein p53 metabolism
- Abstract
Prodelphinidin B-2 3'-O-gallate, a proanthocyanidin gallate isolated from green tea leaf, was investigated for its anti-proliferative activity in human non-small cell lung cancer A549 cells. The results showed that prodelphinidin B-2 3'-O-gallate inhibited the proliferation of A549 cells with no detectable toxic effects on normal WI-38 cells as measured by the XTT assay. Flow cytometric analysis showed that prodelphinidin B-2 3'-O-gallate blocked cell cycle progression in the G0/G1 phase. In addition, prodelphinidin B-2 3'-O-gallate effectively induced A549 cell apoptosis as determined by assessing the nucleosome level in cytoplasm. Enzyme-linked immunosorbent assay showed that the G0/G1 phase arrest is due to p53-independent induction of p21/WAF1. An enhancement in Fas/APO-1 and its two form ligands, membrane-bound Fas ligand (mFasL) and soluble Fas ligand (sFasL), might be responsible for the apoptotic effect induced by prodelphinidin B-2 3'-O-gallate. We suggested that prodelphinidin B-2 3'-O-gallate's activities might be potentially contribute to its overall chemopreventive effects against lung cancer, and can possibly be considered for future therapeutic application.
- Published
- 2005
- Full Text
- View/download PDF
33. Cell cycle stage analysis of rabbit foetal fibroblasts and cumulus cells.
- Author
-
Liu CT, Yu KC, and Ju JC
- Subjects
- Animals, Cell Division, Cells, Cultured, Culture Media, Serum-Free, Female, Flow Cytometry, G1 Phase, Resting Phase, Cell Cycle, Cell Cycle, Fibroblasts cytology, Ovarian Follicle cytology, Rabbits embryology
- Abstract
Synchronization of the cell cycle stages in G0/G1 phase is one of the key factors determining the success of nuclear transplantation. Serum deprivation, contact inhibition and chemical inhibitors are widely used methods for this purpose. In this study, cell cycle stages of foetal fibroblasts and cumulus cells were determined using flow cytometry [fluorescence-activated cell scan (FACS)]. Foetal fibroblasts (in vitro cultured for 72-120 h) and fresh cumulus cells were analysed in Experiment 1. Fifty to 55% proliferating fibroblasts remained in G0/G1 phase compared with 78% in confluent culture (p <0.05). In contrast to foetal fibroblasts, fresh cumulus cells maintained 90% of the population in the G0/G1 stage. When serum was retrieved from the proliferating fibroblasts from day 1 to day 5 (Experiment 2), proportions of G0/G1 cells increased from the initial ratio of 53 to 87% at day 4 of starvation, which was significantly higher than the non-starved proliferating cells (p <0.05). In Experiment 3, fibroblasts were treated with aphidicolin (0.1 microg/ml, 6 h), demicolcine (0.5 microg/ml, 10 h), or a combination of these two chemicals to synchronize the cell cycle stages. Surprisingly, no differences or significantly lower in the proportions of G0/G1)phase cells were detected (25-50%) compared with the uncontrolled growing cells (53%). These results suggested that fresh cumulus cells rest their cell cycle in G0/G1 stage. Serum deprivation became effective in the first 24 h and reached the highest proportions during days 4-5 after deprivation. Chemical synchronization of the cell cycle stage of rabbit foetal fibroblasts to G0/G1 phase appeared less effective compared to serum deprivation.
- Published
- 2004
- Full Text
- View/download PDF
34. Polycomb group gene mel-18 modulates the self-renewal activity and cell cycle status of hematopoietic stem cells.
- Author
-
Kajiume T, Ninomiya Y, Ishihara H, Kanno R, and Kanno M
- Subjects
- Animals, Base Sequence, DNA Primers, DNA-Binding Proteins deficiency, Genes, Homeobox, Hematopoietic Stem Cells cytology, Homeodomain Proteins genetics, Immunosuppression Therapy methods, Mice, Mice, Inbred C57BL, Mice, Knockout, Multigene Family, Polycomb Repressive Complex 1, Pyronine analysis, Resting Phase, Cell Cycle, Reverse Transcriptase Polymerase Chain Reaction, Transcription Factors, Transplantation, Homologous, Whole-Body Irradiation, Zinc Fingers genetics, Bone Marrow Transplantation physiology, Cell Cycle physiology, DNA-Binding Proteins genetics, Hematopoietic Stem Cells physiology
- Abstract
Objective: Mel-18 is a member of the mammalian Polycomb group (PcG) genes. This family of genes regulates global gene expression in many biologic processes, including hematopoiesis and anterior-posterior axis formation by manipulating specific target genes, including members of the Hox family. Here, we demonstrate that mel-18 negatively regulates the self-renewal activity of hematopoietic stem cells (HSCs)., Materials and Methods: Long-term reconstitution activity was evaluated by competitive repopulating unit (CRU) and mean activity of the stem cells (MAS) assays in vivo in bone marrow cells (BMCs) derived from mel-18(-/-) and mel-18 tg mice. The expression levels of mel-18 and Hoxb4 were measured by quantitative real-time reverse transcription polymerase chain reaction., Results: The Hoxb4 gene was highly expressed in HSCs derived from mel-18(-/-) mice. The observed CRUs were 3.21, 4.77, 3.32, and 1.64 CRU per 10(5) BMCs in mel-18(+/+), mel-18(-/-), C57BL/6, and mel-18 tg, respectively. MAS was 0.58, 0.18, 0.41, and 5.89 in mel-18(+/+), mel-18(-/-), C57BL/6, and mel-18 tg, respectively. The percentage in G0 phase HSCs (lin(-)flk2(-)c-Kit(+)Sca1+ cells) was increased in mel-18(-/-) mice and decreased in mel-18 tg mice., Conclusion: Loss or knockdown of mel-18 leads to the expression of Hoxb4, an increase in the proportion of HSCs in G0 phase, and the subsequent promotion of HSC self-renewal. These findings will enable us to develop new approaches for controlling HSC activity for hematopoietic transplantations based on ex vivo expansion of HSCs.
- Published
- 2004
- Full Text
- View/download PDF
35. Cell cycle-dependent accumulation in vivo of transposition-competent complexes between recombination signal ends and full-length RAG proteins.
- Author
-
Jiang H, Ross AE, and Desiderio S
- Subjects
- Animals, Binding Sites, CDC2-CDC28 Kinases metabolism, CHO Cells, Carrier Proteins genetics, Cricetinae, Cyclin-Dependent Kinase 2, DNA-Binding Proteins genetics, G1 Phase, Homeodomain Proteins genetics, Maltose-Binding Proteins, Mice, Mutagenesis, NIH 3T3 Cells, Phosphorylation, Polymerase Chain Reaction, Recombinant Fusion Proteins, Resting Phase, Cell Cycle, Transfection, VDJ Recombinases metabolism, Cell Cycle physiology, DNA-Binding Proteins metabolism, Homeodomain Proteins metabolism, Protein Sorting Signals, Recombination, Genetic, Transposases metabolism
- Abstract
V(D)J recombination is initiated by a specialized transposase consisting of RAG-1 and RAG-2. Because full-length RAG proteins are insoluble under physiologic conditions, most previous analyses of RAG activity in vitro have used truncated core RAG-1 and RAG-2 fragments. These studies identified an intermediate in V(D)J recombination, the signal end complex (SEC), in which core RAG proteins remain associated with recombination signal sequences at the cleaved signal ends. From transfected cells expressing affinity-tagged RAG proteins, we have isolated in vivo assembled SECs containing full-length RAG proteins and cleaved recombination substrates. SEC formation in vivo did not require the repair proteins DNA-dependent protein kinase, Ku80, or XRCC4. In the presence of full-length RAG-2, SEC formation in vivo was cell cycle-regulated and restricted to the G(0)/G(1) phases. In contrast, complexes accumulated throughout cell cycle in cells expressing a RAG-2 CDK2 phosphorylation site mutant. Both core and full-length SECs supported transposition in vitro with similar efficiencies. Intracellular SECs, which are likely to persist in the absence of coding ends, represent potential donors whose transposition is not suppressed by the non-core regions of the RAG proteins.
- Published
- 2004
- Full Text
- View/download PDF
36. Chromosomal changes and cell cycle checkpoints in Mammalian cells.
- Author
-
Geard CR and Ponnaiya B
- Subjects
- Animals, Antimetabolites pharmacology, Bromodeoxyuridine pharmacology, Chromatids ultrastructure, Chromosome Aberrations, Chromosomes ultrastructure, DNA Damage, DNA Repair, Fibroblasts metabolism, G1 Phase, G2 Phase, Humans, In Situ Hybridization, Fluorescence, Metaphase, Mitosis, Resting Phase, Cell Cycle, Ultraviolet Rays, Cell Cycle, Genetic Techniques
- Published
- 2004
- Full Text
- View/download PDF
37. The use of a cell-cycle phase-marker may decrease the percentage of errors when using FISH in PGD.
- Author
-
Pujol A, Benet J, Campillo M, Codina-Pascual M, Egozcue J, and Navarro J
- Subjects
- Adult, Cells, Cultured, Chromatin, Chromosomes, Human, Congenital Abnormalities diagnosis, Congenital Abnormalities genetics, Genetic Testing, Genotype, Humans, Male, Reproducibility of Results, Resting Phase, Cell Cycle, Cell Cycle, DNA Probes, In Situ Hybridization, Fluorescence methods, Lymphocytes cytology, Sertoli Cells cytology
- Abstract
Fluorescent DNA probes are used to characterise the chromosome constitution of preimplantation embryos. FISH is used to select normal or balanced embryos in carriers of balanced chromosomal rearrangements, for embryo sexing or for aneuploidy screening in women of advanced age, who have had recurrent abortions or IVF failures. In most cases, FISH is performed on interphase blastomeres which are asynchronously dividing cells, that can be in G1, S or G2. However, a correct interpretation of a double FISH signal, which may correspond to a split signal, to a replicated chromosome region or to the presence of an extra chromosome is essential to establish an accurate diagnosis. To determine if the cell stage could influence the interpretation of FISH results, we compared the signal characteristics of one locus-specific probe, two different subtelomere region probes, and a centromere region probe in non-dividing Sertoli cells and in proliferating lymphocytes. Most cells had two signals per chromosome pair (i.e., a situation corresponding to G0 in Sertoli cells and to G1 or to a prereplication stage in lymphocytes). Nevertheless, in proliferating cells the percentage of nuclei with a number of signals different from the expected (two unreplicated chromosomes per pair) was different from that found in non-dividing cells (P < 0.05). It was estimated that 10.8% of double dots in dividing cells resulted from DNA replication. The sequence of replication was first the locus-specific region, second a telomere region, and third the centromere. In conclusion, the DNA replication process could result in errors of interpretation (misdiagnosis) in 7% of proliferating cells. Thus, the use of a cell cycle phase-specific marker could avoid errors by indicating the cell stage in which the nucleus analysed is found., (Copyright 2003 S. Karger AG, Basel)
- Published
- 2004
- Full Text
- View/download PDF
38. G0-G1 cell cycle phase transition as revealed by fluorescence resonance energy transfer: analysis of human fibroblast chromatin.
- Author
-
Bottiroli G, Croce AC, Bottone MG, Vaccino S, and Pellicciari C
- Subjects
- Binding Sites, Cells, Cultured, Chromatin metabolism, Cytokines metabolism, DNA metabolism, Fibroblasts cytology, Fibroblasts metabolism, G1 Phase, Humans, Resting Phase, Cell Cycle, Spectrometry, Fluorescence methods, Staining and Labeling, Cell Cycle physiology, Chromatin chemistry, Fibroblasts chemistry, Fluorescence Resonance Energy Transfer methods
- Abstract
In the present study, microspectrofluorometry and digital imaging procedures were used to investigate by fluorescence Resonance Energy Transfer (FRET) analysis the changes of chromatin organization during the transition from G0 quiescent stat to G1 phase. G0 transition is a key event in cell cycle progress depending on the activation of specific genes and the concomitant silencing of others, which both entail spatial chromatin rearrangement. Normal human fibroblasts arrested in G0-phase by culture in low-serum containing medium and stimulated to re-enter G1 by serum addition were used as cell model. To investigate the occurrence and timing of these supramolecular chromatin changes, we estimated the relative FRET efficiency in single cells after double-helical DNA. Hoechst 33258 amd propidium iodide were used as a donor-acceptor dye pair since they exhibit particularly favourable spectral characteristics, that allow the calculation procedure to be semplified. The results of FRET analysis were compared to those of the immunocytochemical labelling of two nuclear proteins (i.e., Ki-67 and statin) whose expression is an established marker of potentially proliferating G1 cells or resting G0 cells, respectively. FRET efficiency was lower in G0 than G1 fibroblasts: this is likely due to higher chromatin packaging in quiescent cells which especially hinders the interaction with the donor molecules less favourable, in terms of relative distance and spatial orientation. FRET efficiency significantly increased shortly (1h) after serum stimulation of quiescent fibroblasts, thus indicating that chromatin is rearranged in parallel with activation of cycle-related gene; it is worth noting that these signs largely preceded the occurrence of immunopositivity for Ki-67, which was detectable only 24h after serum stimulation. FRET-based analyses which already proved to be suitable for studying the overall chromatin organization in differentiated cells, may now be envisaged as a powerful tool for detecting, in single cells, more subtle changes linked to the activation of early cycle-related genes.
- Published
- 2004
39. Cell differentiation and proliferation--simultaneous but independent?
- Author
-
Brown G, Hughes PJ, and Michell RH
- Subjects
- Animals, Cell Division, Humans, Resting Phase, Cell Cycle, Cell Cycle, Cell Differentiation
- Abstract
Despite studies over many years, it is still not clear to what extent cellular controls on proliferation and on differentiation are interrelated. For example, the idea that exit from cell cycle into G1/G0 is a necessary-or at least frequent-prelude to differentiation developed partly from studies of haemopoietic cell maturation, often using cell lines as models. The responses of cells to treatment with differentiating agents suggested that exit from cell cycle into G1/G0 occurs quite quickly, with functional differentiated characteristics acquired later, and so promoted the notion that cyclin-dependent kinase inhibitors (CDKIs) might be important initiators of normal differentiation. However, recent work has made it clear that differentiation and cell proliferation are regulated simultaneously but independently, that cells often start differentiating long before they stop dividing, and that the launching of differentiation is not restricted to any particular segment of the cell cycle. This combination of attributes allows expansion of cell numbers and acquisition of differentiated function to occur in parallel, generating abundant effector cells. Given this dichotomy, future studies to develop a more exact picture of the events that initiate and drive differentiation might be simplified by studying differentiation under experimental conditions that divorce it from concerns about cell cycle control.
- Published
- 2003
- Full Text
- View/download PDF
40. Bcl-xL/Bcl-2 coordinately regulates apoptosis, cell cycle arrest and cell cycle entry.
- Author
-
Janumyan YM, Sansam CG, Chattopadhyay A, Cheng N, Soucie EL, Penn LZ, Andrews D, Knudson CM, and Yang E
- Subjects
- Alleles, Animals, Carrier Proteins metabolism, Cell Division, Cell Line, Contact Inhibition, G1 Phase, Kinetics, Mutagenesis, Proto-Oncogene Proteins c-bcl-2 genetics, Rats, Recombinant Proteins metabolism, Resting Phase, Cell Cycle, bcl-Associated Death Protein, bcl-X Protein, Apoptosis physiology, Cell Cycle physiology, Proto-Oncogene Proteins c-bcl-2 metabolism
- Abstract
Bcl-x(L) and Bcl-2 inhibit both apoptosis and proliferation. In investigating the relationship between these two functions of Bcl-x(L) and Bcl-2, an analysis of 24 Bcl-x(L) and Bcl-2 mutant alleles, including substitutions at residue Y28 previously reported to selectively abolish the cell cycle activity, showed that cell cycle delay and anti-apoptosis co-segregated in all cases. In determining whether Bcl-2 and Bcl-x(L) act in G(0) or G(1), forward scatter and pyronin Y fluorescence measurements indicated that Bcl-2 and Bcl-x(L) cells arrested more effectively in G(0) than controls, and were delayed in G(0)-G(1) transition. The cell cycle effects of Bcl-2 and Bcl-x(L) were reversed by Bad, a molecule that counters the survival function of Bcl-2 and Bcl-x(L). When control and Bcl-x(L) cells of equivalent size and pyronin Y fluorescence were compared, the kinetics of cell cycle entry were similar, demonstrating that the ability of Bcl-x(L) and Bcl-2 cells to enhance G(0) arrest contributes significantly to cell cycle delay. Our data suggest that cell cycle effects and increased survival both result from intrinsic functions of Bcl-2 and Bcl-x(L).
- Published
- 2003
- Full Text
- View/download PDF
41. Regulation of bovine corneal endothelial cell cycle by transforming growth factor-beta.
- Author
-
Motegi Y, Usui T, Ishida K, Kato S, and Yamashita H
- Subjects
- Animals, Antigens, CD, Cattle, Cell Cycle physiology, Cells, Cultured, Endoglin, Endothelium, Corneal metabolism, G1 Phase, Humans, Protein Kinase C metabolism, Proto-Oncogene Proteins c-sis biosynthesis, Receptors, Cell Surface, Receptors, Transforming Growth Factor beta metabolism, Resting Phase, Cell Cycle, S Phase, Signal Transduction physiology, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta1, Transforming Growth Factor beta2, Vascular Cell Adhesion Molecule-1 metabolism, Cell Cycle drug effects, Endothelium, Corneal cytology, Transforming Growth Factor beta pharmacology
- Abstract
Purpose: The transforming growth factor-beta (TGF-beta) family includes three multifunctional proteins, TGF-beta1, TGF-beta2 and TGF-beta3, expressed in ocular tissue, which are involved in regulating cell differentiation, cell proliferation and other cell functions. TGF-beta is present in aqueous humour and regulates corneal endothelial cells. This study explores the mechanism by which TGF-beta regulates the cell cycle in cultured corneal endothelial cells., Methods: The expression of specific receptors for the TGF-beta family was investigated at the protein level by affinity cross-linking with radio-iodinated TGF-beta1 and immunoprecipitation with specific antibodies to TGF-beta receptors. Regulation of entry into the S-phase of the cell cycle was determined by 5-bromo-2' deoxyuridine (BrdU) incorporation into the cells. The signal transduction pathways were investigated using various blocking agents for protein kinase transducers involved in intracytoplasmic signal transduction., Results: Cultured bovine corneal endothelial cells were confirmed to express TGF-beta type 1 and type 2 receptors and endoglin. In the confluent state, TGF-beta1 and TGF-beta2 stimulated the cells to progress to the S-phase of the cell cycle through platelet-derived growth factor-B (PDGF-B) chain production and protein kinase C., Conclusions: TGF-beta accelerated cell cycle progression from the G0/G1 phase to the S-phase in cultured corneal endothelial cells, under our experimental conditions, through pathways involving protein kinase C. These pathways are related to the cross-talk between TGF-beta and other cytokines. The conditions employed in the present experiments may be useful for investigating the complex cross-talk between various cytokines and growth factors.
- Published
- 2003
- Full Text
- View/download PDF
42. Heme oxygenase-1 expression levels are cell cycle dependent.
- Author
-
Colombrita C, Lombardo G, Scapagnini G, and Abraham NG
- Subjects
- Anions, Bilirubin metabolism, Blotting, Western, Carbon Monoxide metabolism, Cell Division, Cells, Cultured, Culture Media, Serum-Free pharmacology, DNA Damage, Endothelium, Vascular metabolism, F2-Isoprostanes biosynthesis, G1 Phase, Heme Oxygenase-1, Humans, Membrane Proteins, Mesoporphyrins pharmacology, Microscopy, Confocal, Oxidative Stress, Oxygen metabolism, Resting Phase, Cell Cycle, Reverse Transcriptase Polymerase Chain Reaction, Superoxides metabolism, Temperature, Time Factors, Up-Regulation, Cell Cycle, Dinoprost analogs & derivatives, Heme Oxygenase (Decyclizing) biosynthesis
- Abstract
Heme oxygenase-1 (HO-1) is a stress protein, which has been suggested to participate in defense mechanisms against agents that may induce oxidative injury, such as angiotensin II (Ang II). The purpose of the present study was to examine the role of human HO-1 in cell-cycle progression. We investigated the effect of Ang II on HO-1 gene expression in serum-deprived media to drive human endothelial cells into G(0)/G(1) (1% FBS) compared to exponentially grown cells (10% FBS). The addition of Ang II (100 ng/ml) to endothelial cells increased HO-1 protein and activity in G(0)/G(1) in a time-dependent manner, reaching a maximum HO-1 level at 16 h. Real-time RT-PCR demonstrated that Ang II increased the levels of HO-1 mRNA in G(0)/G(1) as early as 1 h. The rate of HO-1 induction in response to Ang II was several-fold higher in serum-starved cells compared to cells cultured in continuous 10% FBS. The addition of Ang II increased the generation of 8-epi-isoprostane PGF(2 alpha). Inhibition of HO-1, by Stannis mesoporphyrin (SnMP), potentiated Ang II-mediated DNA damage and generation of 8-epi-isoprostane PGF(2 alpha). These results imply that expression of HO-1 in G(0)/G(1), in the presence of Ang II, may be a key player in attenuating DNA damage during cell-cycle progression. Thus, exposure of endothelial cells to Ang II causes a complex response involving generation of superoxide anion, which may be involved in DNA damage. Upregulation of HO-1 ensures the generation of bilirubin and carbon monoxide (CO) in G(0)/G(1) phase to counteract Ang II-mediated oxidative DNA damage. Inducibility of HO-1 in G(0)/G(1) phase is essential and probably regulated by a complex system involving oxygen species to assure controlled cell growth.
- Published
- 2003
- Full Text
- View/download PDF
43. P21Waf1/Cip1 dysfunction in neuroblastoma: a novel mechanism of attenuating G0-G1 cell cycle arrest.
- Author
-
McKenzie PP, Danks MK, Kriwacki RW, and Harris LC
- Subjects
- Child, Cyclin-Dependent Kinase 2, Cyclin-Dependent Kinase 4, Cyclin-Dependent Kinase Inhibitor p21, Cyclin-Dependent Kinases metabolism, Cyclins genetics, G1 Phase, Genetic Vectors, Humans, Protein Serine-Threonine Kinases metabolism, Recombinant Proteins metabolism, Resting Phase, Cell Cycle, Transfection, CDC2-CDC28 Kinases, Cell Cycle physiology, Cyclins metabolism, Neuroblastoma pathology, Proto-Oncogene Proteins
- Abstract
In normal cells in which DNA has been damaged, p53 induces the expression of p21(Waf1/Cip1); p21, in turn, binds to cyclin-dependent kinase 2 (cdk2) and inhibits its function. Inhibition of cdk2 results in cell cycle arrest in G(0)-G(1). Although p53 is transcriptionally active and induces p21 expression in neuroblastoma (NB) cells, the G(0)-G(1) checkpoint is attenuated. Here we report that the mechanism that mediates this defect in NB cells is the inability of p21 to bind to, or inhibit the activity of cdk2. However, when recombinant p21 protein was added to NB cell extracts in vitro, the protein inhibited the activity of cdk2. This finding suggests that endogenous p21 protein in NB cells is inactive and may be bound either to a protein complex or in a conformation that precludes its binding to cdk2. The dysfunction of p21 in NB cells represents a novel mechanism by which the G(0)-G(1) cell cycle checkpoint can be inactivated. This mechanism may be important in regulating the growth of NB and potentially other types of tumors. Cdk inhibitors currently being developed for clinical use may be useful therapy for tumors such as NB in which endogenous cdk inhibitors are defective.
- Published
- 2003
44. Are chromosomal instabilities induced by exposure of cultured normal human cells to low- or high-LET radiation?
- Author
-
Dugan LC and Bedford JS
- Subjects
- Cell Division radiation effects, Cells, Cultured, Cesium Radioisotopes, Fibroblasts metabolism, Humans, Hydrogen-Ion Concentration, Image Processing, Computer-Assisted, In Situ Hybridization, Fluorescence, Iron, Metaphase radiation effects, Mitosis, Radiation Tolerance, Resting Phase, Cell Cycle, Time Factors, Cell Cycle radiation effects, Chromosome Aberrations, Chromosomes radiation effects
- Abstract
Radiation-induced genomic instability has been proposed as a very early, if not an initiating, step in radiation carcinogenesis. Numerous studies have established the occurrence of radiation-induced chromosomal instability in various cells of both human and rodent origin. In many of these studies, however, the cells were not "normal" initially, and in many cases they involved tumor-derived cell lines. The phenomenon clearly would be of even greater interest if it were shown to occur generally in cells that are normal at the outset, rather than cells that may have been "selected" because of a pre-existing susceptibility to induced instability. As a test of the generality of the phenomenon, we studied low-passage normal diploid human fibroblasts (AG1521A) to determine whether they are susceptible to the induction of chromosomal instability in the progeny of surviving cells after exposure in G(0) to low- and high-LET radiation. Cytogenetic assays for instability were performed on both mixed populations of cells and clones of cells surviving exposure. We found no evidence for the induction of such instability as a result of radiation exposure, though we observed a senescence-related chromosomal instability in the progeny of both irradiated and unirradiated cell populations., (Copyright 2003 by Radiation Research Society)
- Published
- 2003
- Full Text
- View/download PDF
45. Coordination between donor cell type and cell cycle stage improves nuclear cloning efficiency in cattle.
- Author
-
Wells DN, Laible G, Tucker FC, Miller AL, Oliver JE, Xiang T, Forsyth JT, Berg MC, Cockrem K, L'Huillier PJ, Tervit HR, and Oback B
- Subjects
- Animals, Cattle embryology, Embryo Transfer veterinary, Embryonic and Fetal Development, Female, Fibroblasts ultrastructure, G1 Phase, G2 Phase, Mitosis, Pregnancy, Resting Phase, Cell Cycle, Cattle genetics, Cell Cycle, Cloning, Organism, Nuclear Transfer Techniques
- Abstract
Several studies have shown that both quiescent and proliferating somatic donor cells can be fully reprogrammed after nuclear transfer (NT) and result in viable offspring. So far, however, no comparative study has conclusively demonstrated the relative importance of donor cell cycle stage on nuclear cloning efficiency. Here, we compare two different types of bovine fetal fibroblasts (BFFs) that were synchronized in G(0), G(1), and different phases within G(1). We show that for non-transgenic (non-TG) fibroblasts, serum starvation into G(0) results in a significantly higher percentage of viable calves at term than synchronization in early G(1) or late G(1). For transgenic fibroblasts, however, cells selected in G(1) show significantly higher development to calves at term and higher post-natal survival to weaning than cells in G(0). This suggests that it may be necessary to coordinate donor cell type and cell cycle stage to maximize overall cloning efficiency., (Copyright 2002 Elsevier Science Inc.)
- Published
- 2003
- Full Text
- View/download PDF
46. Flow cytometric cell cycle analysis of somatic cells primary cultures established for bovine cloning.
- Author
-
Katska L, Bochenek M, Kania G, Ryñska B, and Smorag Z
- Subjects
- Animals, Cells, Cultured, Cryopreservation, Culture Media, Serum-Free, Female, G1 Phase, G2 Phase, Mitosis, Oocytes cytology, Resting Phase, Cell Cycle, Cattle, Cell Cycle, Cloning, Organism, Fibroblasts, Flow Cytometry, Ovarian Follicle cytology
- Abstract
An important factor governing developmental rates of somatic cloned embryos is the phase of the cell cycle of donor nuclei. The aim of this experiment was to investigate the distribution of cell cycle phases in bovine cumulus and fibroblast cells cultured using routine treatment, and under cell cycle-arresting treatments. The highest percentages of cumulus cells in the G0 + G1 stage were observed in uncultured, frozen/thawed cells originating from immature oocytes (79.8 +/- 2.2%), fresh and frozen/thawed cells from in vitro matured oocytes (84.1 +/- 6.2 and 77.8 +/- 5.7%, respectively), and in cycling cells (72.7 +/- 16.3 and 78.4 +/- 11.2%, respectively for cumulus cells from immature and in vitro matured oocytes). Serum starvation of cumulus cultures markedly decreased percentages of cells in G0 + G1, and prolonged starvation significantly increased (P < 0.05) percentages of cells in G2 + M phase. Culture of cumulus cells to confluency did not increase percentages of cells in G0 + G1. Contrary to findings in cumulus cells, significantly higher percentages of cells in G0 + G1 were apparent when fibroblast cells were cultured to confluency or serum starved, and significantly increased (P < 0.01) as the starvation period was prolonged. It is concluded that for particular cell types specific strategies should be used to attain improvements in the efficiency of cloning procedures.
- Published
- 2002
- Full Text
- View/download PDF
47. Activation of cyclin D1-kinase in murine fibroblasts lacking both p21(Cip1) and p27(Kip1).
- Author
-
Sugimoto M, Martin N, Wilks DP, Tamai K, Huot TJ, Pantoja C, Okumura K, Serrano M, and Hara E
- Subjects
- Animals, Cattle, Cell Cycle drug effects, Cell Cycle Proteins physiology, Contact Inhibition, Culture Media pharmacology, Culture Media, Serum-Free, Cyclin-Dependent Kinase 4, Cyclin-Dependent Kinase Inhibitor p21, Cyclin-Dependent Kinase Inhibitor p27, Cyclins physiology, Embryo, Mammalian cytology, Enzyme Activation drug effects, Fetal Blood physiology, Fibroblasts cytology, Fibroblasts drug effects, Gene Targeting, Growth Substances pharmacology, Humans, Macromolecular Substances, Mice, Mitogens pharmacology, Resting Phase, Cell Cycle, S Phase, Trans-Activators physiology, Transcription Factors, Tumor Suppressor Proteins physiology, Cell Cycle physiology, Cyclin-Dependent Kinases metabolism, Cyclins deficiency, Fibroblasts metabolism, Nuclear Proteins, Proto-Oncogene Proteins, Tumor Suppressor Proteins deficiency
- Abstract
Deregulation of D-type cyclin-dependent kinases (CDK4 and 6) is widely observed in various human cancers, illustrating their importance in cell cycle control. Like other cyclin-dependent kinases (CDKs), assembly with cyclins is the most critical step for activation of CDK4/6. As previously reported elsewhere, we observed that the level of cyclinD1-CDK4 complex and its associated kinase activity were significantly low in asynchronously proliferating mouse embryo fibroblasts lacking both p21(Cip1) and p27(Kip1) (p21/p27-null MEFs). These evidences imply that p21(Cip1) and p27(Kip1) CDK inhibitors are 'essential activators' of cyclin D-kinases. We, however, discovered here that both the assembly and activation of cyclin D1-CDK4 complex occur when quiescent p21/p27-null MEFs were stimulated to re-enter the cell cycle. This mitogen-induced cyclin D1-kinase activity was blocked by overexpression of p16(INK4a) and resulted in the inhibition of S phase entry in p21/p27-null MEFs. Furthermore, ectopic expression of p34(SEI-1), a mitogen-induced CDK4 binding protein, increased the levels of active cyclinD1-CDK4 complex in asynchronously proliferating p21/p27-null MEFs. Together, our results suggest that there are several independent ways to stimulate the assembly of cyclin D1-CDK4 kinases. Although p21(Cip1) and p27(Kip1) play a role in this process, our results demonstrate that additional mechanisms must occur in G0 to S phase transition.
- Published
- 2002
- Full Text
- View/download PDF
48. A role for p53 in maintaining and establishing the quiescence growth arrest in human cells.
- Author
-
Itahana K, Dimri GP, Hara E, Itahana Y, Zou Y, Desprez PY, and Campisi J
- Subjects
- Antigens, Viral, Tumor genetics, Antigens, Viral, Tumor metabolism, Cells, Cultured, DNA Replication, Electrophoresis, Polyacrylamide Gel, Fibroblasts cytology, Humans, Microinjections, RNA, Messenger metabolism, Resting Phase, Cell Cycle, Transcriptional Activation, Cell Cycle physiology, Tumor Suppressor Protein p53 physiology
- Abstract
The p53 tumor suppressor protein induces transient growth arrest or apoptosis in response to genotoxic stress and mediates the irreversible growth arrest of cellular senescence. We present evidence here that p53 also contributes to the reversible, growth factor-dependent arrest of quiescence (G(0)). Microinjection of expression vectors encoding either MDM2 or a pRb-binding mutant of SV40 T antigen, both of which abrogate p53 function, stimulated quiescent normal human fibroblasts to initiate DNA synthesis and were 40-70% as effective as wild-type T antigen. Electrophoretic mobility shift and p53 transactivation assays showed that p53 activity was higher in quiescent and senescent cells compared with proliferating cells. As proliferating cells entered G(0) after growth factor withdrawal, the p53 mRNA level increased, followed by transient accumulation of the protein. Shortly thereafter, the expression (mRNA and protein) of p21, a p53 target gene and effector of cell cycle arrest, increased. Finally, stable expression of the HPV16 E6 oncogene or dominant negative p53 peptide, GSE-22, both of which inhibit p53 function, delayed entry into quiescence following growth factor withdrawal. Our data indicate that p53 is activated during both quiescence and senescence. They further suggest that p53 activity contributes, albeit not exclusively, to the quiescent growth arrest.
- Published
- 2002
- Full Text
- View/download PDF
49. Differential expression of receptors for Shiga and Cholera toxin is regulated by the cell cycle.
- Author
-
Majoul I, Schmidt T, Pomasanova M, Boutkevich E, Kozlov Y, and Söling HD
- Subjects
- Animals, Cells, Cultured, Chlorocebus aethiops, G(M1) Ganglioside biosynthesis, G1 Phase, G2 Phase, Hippocampus cytology, Mice, Neurons metabolism, PC12 Cells, Rats, Receptors, Cell Surface biosynthesis, Resting Phase, Cell Cycle, Telophase, Trihexosylceramides biosynthesis, Vero Cells, Cell Cycle, G(M1) Ganglioside metabolism, Receptors, Cell Surface metabolism, Trihexosylceramides metabolism
- Abstract
Cholera and Shiga toxin bind to the cell surface via glycolipid receptors GM1 and Gb3, respectively. Surprisingly, the majority of Vero cells from a non-synchronized population bind either Cholera or Shiga toxin but not both toxins. The hypothesis that the differential expression of toxin receptors is regulated by the cell cycle was tested. We find that Cholera toxin binds preferentially in G0/G1, with little binding through S-phase to telophase, whereas Shiga toxin binds maximally through G2 to telophase but does not bind during G0/G1 and S-phase. The changes result from the corresponding changes in Gb3 and GM1 synthesis, not from variations of receptor transport to the cell surface. The changes do not reflect competition of Gb3 and GM1 synthesis for lactosylceramide. Cells as diverse as Vero cells, PC12 cells and astrocytes show the same cell-cycle-dependent regulation of glycosphingolipid receptors, suggesting that this novel phenomenon is based on a conserved regulatory mechanism.
- Published
- 2002
- Full Text
- View/download PDF
50. Regulation of Ras signaling by the cell cycle.
- Author
-
Stacey D and Kazlauskas A
- Subjects
- Animals, Cyclin D1 metabolism, G1 Phase, Gene Expression Regulation, Humans, Phosphatidylinositol 3-Kinases physiology, Proto-Oncogene Proteins c-raf physiology, Resting Phase, Cell Cycle, S Phase, ral Guanine Nucleotide Exchange Factor physiology, ras Proteins metabolism, Cell Cycle physiology, Signal Transduction physiology, ras Proteins physiology
- Abstract
It is well known that upregulation of Ras activity can promote cell-cycle progression. Now recent studies indicate that a reciprocal relationship also exists; that is, the consequences of Ras signaling are dependent upon cell-cycle position. In quiescent cells stimulated with growth factors, one Ras effector, phosphatidylinositol-3-kinase, is activated twice as cells transition from G(0) into G(1) phase, and then later in G(1) phase. It is only during the later stages of G(1) phase that PI3K activity promotes entry into S-phase. In cycling cells, Ras activity is enhanced throughout the cell cycle, but is able to stimulate cyclin D1 elevation only during G(2) phase.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.