• Title/Summary/Keyword: cell cycle regulation

Search Result 489, Processing Time 0.022 seconds

Cell Cycle Regulation in the Budding Yeast

  • Nguyen, Cuong;Yoon, Chang-No;Han, Seung-Kee
    • Proceedings of the Korean Society for Bioinformatics Conference
    • /
    • 2005.09a
    • /
    • pp.278-283
    • /
    • 2005
  • Cell cycle is regulated cooperatively by several genes. The dynamic regulatory mechanism of protein interaction network of cell cycle will be presented taking the budding yeast as a sample system. Based on the mathematical model developed by Chen et at. (MBC, 11,369), at first, the dynamic role of the feedback loops is investigated. Secondly, using a bifurcation diagram, dynamic analysis of the cell cycle regulation is illustrated. The bifurcation diagram is a kind of ‘dynamic road map’ with stable and unstable solutions. On the map, a stable solution denotes a ‘road’ attracting the state and an unstable solution ‘a repelling road’ The ‘START’ transition, the initiation of the cell cycle, occurs at the point where the dynamic road changes from a fixed point to an oscillatory solution. The 'FINISH' transition, the completion of a cell cycle, is returning back to the initial state. The bifurcation analysis for the mutants could be used uncovering the role of proteins in the cell cycle regulation network.

  • PDF

PP2A function toward mitotic kinases and substrates during the cell cycle

  • Jeong, Ae Lee;Yang, Young
    • BMB Reports
    • /
    • v.46 no.6
    • /
    • pp.289-294
    • /
    • 2013
  • To maintain cellular homeostasis against the demands of the extracellular environment, a precise regulation of kinases and phosphatases is essential. In cell cycle regulation mechanisms, activation of the cyclin-dependent kinase (CDK1) and cyclin B complex (CDK1:cyclin B) causes a remarkable change in protein phosphorylation. Activation of CDK1:cyclin B is regulated by two auto-amplification loops-CDK1:cyclin B activates Cdc25, its own activating phosphatase, and inhibits Wee1, its own inhibiting kinase. Recent biological evidence has revealed that the inhibition of its counteracting phosphatase activity also occurs, and it is parallel to CDK1:cyclin B activation during mitosis. Phosphatase regulation of mitotic kinases and their substrates is essential to ensure that the progression of the cell cycle is ordered. Outlining how the mutual control of kinases and phosphatases governs the localization and timing of cell division will give us a new understanding about cell cycle regulation.

Apoptotic effect of $IP_6$ was not enhanced by co-treatment with myo-inositol in prostate carcinoma PC3 cells

  • Kim, Hyun-Jung;Jang, Yu-Mi;Kim, Harriet;Kwon, Young-Hye
    • Nutrition Research and Practice
    • /
    • v.1 no.3
    • /
    • pp.195-199
    • /
    • 2007
  • Inositol hexaphosphate ($IP_6$) is a major constituent of most cereals, legumes, nuts, oil seeds and soybean. Previous studies reported the anticancer effect of $IP_6$ and suggested that co-treatment of $IP_6$ with inositol may enhance anticancer effect of $IP_6$. Although the anticancer effect of $IP_6$ has been intensively studied, the combinational effect of $IP_6$ and inositol and involved mechanisms are not well understood so far. In the present study, we investigated the effect of $IP_6$ and myo-inositol (MI) on cell cycle regulation and apoptosis using PC3 prostate cancer cell lines. When cell, were co-treated with $IP_6$ and MI, the extent of cell growth inhibition was significantly increased than that by $IP_6$ alone. To identify the effect of $IP_6$ and MI on apoptosis, the activity of caspase-3 was measured. The caspase-3 activity was significantly increased when cells were treated with either $IP_6$ alone or both $IP_6$ and MI, with no significant enhancement by co-treatment. To investigate the effect of $IP_6$ and MI of cell cycle arrest, we measured p21 mRNA expression in PC3 cells and observed significant increase in p21 mRNA by $IP_6$. But synergistic regulation by co-treatment with $IP_6$ and MI was not observed. In addition, there was no significant effect by co-treatment compared to $IP_6$ treatment on the regulation of cell cycle progression although $IP_6$ significantly changed cell cycle distribution in the presence of MI or not. Therefore, these findings support that $IP_6$ has anticancer function by induction of apoptosis and regulation of cell cycle. However, synergistic effect by MI on cell cycle regulation and apoptosis was not observed in PC3 prostate cancer cells.

Retinoic Acid Increases the Cell Cycle Progression of Human Gingival Fibroblasts by Increasing Cyclin E and CDK 2 Expression and Decreasing $p21^{WAF1/CIP1}$ and $p16^{INK4A}$ Expression

  • You, Hyung-Keun;Seo, Se-Jeong;Kim, Kang-Ju;Choi, Na-Young;You, Yong-Ouk
    • International Journal of Oral Biology
    • /
    • v.37 no.3
    • /
    • pp.115-120
    • /
    • 2012
  • Retinoic acid plays an important role in the regulation of cell growth and differentiation. In our present study, we evaluated the effects of all-trans retinoic acid (RA) on cell proliferation and on the cell cycle regulation of human gingival fibroblasts (HGFs). Cell proliferation was assessed using the MTT assay. Cell cycle analysis was performed by flow cytometry, and cell cycle regulatory proteins were determined by western blot. Cell proliferation was increased in the presence of a 0.1 nM to 1 ${\mu}M$ RA dose range, and maximal growth stimulation was observed in cells exposed to 1 nM of RA. Exposure of HGFs to 1 nM of RA resulted in an augmented cell cycle progression. To elucidate the molecular mechanisms underlying cell cycle regulation by RA, we measured the intracellular levels of major cell cycle regulatory proteins. The levels of cyclin E and cyclin-dependent kinase (CDK) 2 were found to be increased in HGFs following 1 nM of RA treatment. However, the levels of cyclin D, CDK 4, and CDK 6 were unchanged under these conditions. Also after exposure to 1 nM of RA, the protein levels of $p21^{WAF1/CIP1}$ and $p16^{INK4A}$ were decreased in HGFs compared with the control group, but the levels of p53 and pRb were similar between treated and untreated cells. These results suggest that RA increases cell proliferation and cell cycle progression in HGFs via increased cellular levels of cyclin E and CDK 2, and decreased cellular levels of $p21^{WAF1/CIP1}$ and $p16^{INK4A}$.

Oct4 resetting by Aurkb–PP1 cell cycle axis determines the identity of mouse embryonic stem cells

  • Shin, Jihoon;Youn, Hong-Duk
    • BMB Reports
    • /
    • v.49 no.10
    • /
    • pp.527-528
    • /
    • 2016
  • In embryonic stem cells (ESCs), cell cycle regulation is deeply connected to pluripotency. Especially, core transcription factors (CTFs) which are essential to maintaining the pluripotency transcription programs should be reset during M/G1 transition. However, it remains unknown about how CTFs are governed during cell cycle progression. Here, we describe that the regulation of Oct4 by Aurora kinase b (Aurkb)/protein phosphatase 1 (PP1) axis during the cell cycle is important for resetting Oct4 to pluripotency and cell cycle related target genes in determining the identity of ESCs. Aurkb starts to phosphorylate Oct4(S229) at the onset of G2/M phase, inducing the dissociation of Oct4 from chromatin, whereas PP1 binds Oct4 and dephosphorylates Oct4(S229) during M/G1 transition, which resets Oct4-driven transcription for pluripotency and the cell cycle. Furthermore, Aurkb phosphormimetic and PP1 binding-deficient mutations in Oct4 disrupt the pluripotent cell cycle, lead to the loss of pluripotency in ESCs, and decrease the efficiency of somatic cell reprogramming. Based on our findings, we suggest that the cell cycle is directly linked to pluripotency programs in ESCs.

Bifurcation analysis of budding yeast cell cycle

  • Nguyen, Cuong;Yoon, Chang-No;Kim, Hak-Yong;Han, Seung-Kee
    • Proceedings of the Korean Society for Bioinformatics Conference
    • /
    • 2004.11a
    • /
    • pp.50-56
    • /
    • 2004
  • Bifurcation analysis of cell cycle regulation in the budding yeast is performed basedon the mathematical model by Chen et al [Molecular biology of cell, 11:369-391, 2000]. On the bifurcation diagram, locations of both stable and unstable solutions of the nonlinear differential equations are presented by taking the mass of cell as a controlparameter. Based on the bifurcation diagram, dynamic mechanism underlying the 'start' transition, initiation of a new round of cell cycle, and the 'finish' transition, completion of cell cycle and returning back to the initial state, is discussed: the 'start' transition is a transition from a stable fixed solution for a small mass and to an oscillatory state for a large mass, and the 'finish' transition is a switching back to the stable fixed solution from the oscillatory state. To understand the role of the genes during the cell cycle regulation, bifurcation diagrams for the mutants are compared with that of the wild type.

  • PDF

Linear Dynamic Model of Gene Regulation Network of Yeast Cell Cycle

  • Changno Yoon;Han, Seung-Kee
    • Proceedings of the Korean Biophysical Society Conference
    • /
    • 2003.06a
    • /
    • pp.77-77
    • /
    • 2003
  • Gene expression in a cell is regulated by mutual activations or repressions between genes. Identifying the gene regulation network will be one of the most important research topics in the post genomic era. We propose a linear dynamic model of gene regulation for the yeast cell cycle. A small gene network consisting of about 40 genes is reconstructed from the analysis of micro-array gene expression data of yeast S. cerevisiae published by P. Spellman et al. We show that the network construction is consistent with the result of the hierarchical cluster analysis.

  • PDF

Role of the Promoter Region of a Chicken H3 Histone Gene in Its Cell Cycle Dependent Expression

  • Son, Seung-Yeol
    • BMB Reports
    • /
    • v.32 no.4
    • /
    • pp.345-349
    • /
    • 1999
  • We fused the promoter region of an H3.2 chicken histone gene, whose expression is dependent on the cell cycle, to the 5' coding region of an H3.3 chicken histone gene, which is expressed constitutively at a low level throughout the cell cycle. This fusion gene showed a cell cycle-regulated pattern of expression, but in a different manner. The mRNA level of the fusion gene increase during the S phase of the cell cycle by about 3.7-fold at 6 h and 2.7-fold at 12 h after the serum stimulation. The mRNA level of the intact H3.2 gene, however, increased by an average of 3.6-fold at 6 h and 8.7-fold at 12 h. This different expression pattern might be due to the differences in their 3' end region that is responsible for mRNA stability. The 3' end of the H3.2 mRNA contains a stem-loop structure, instead of a poly(A) tail present in the H3.3 mRNA. We also constructed a similar fusion gene using a H3.3 histone gene whose introns had been eliminated to rule out the possibility of involvement of the introns in cell cycle-regulated expression. The expression of this fusion gene was almost identical to the fusion gene made previously. These results indicate that the promoter region of the H3.2 gene is only partially responsible for its expression during the S phase of the cell cycle.

  • PDF

Down-regulation of the cyclin E1 oncogene expression by microRNA-16-1 induces cell cycle arrest in human cancer cells

  • Wang, Fu;Fu, Xiang-Dong;Zhou, Yu;Zhang, Yi
    • BMB Reports
    • /
    • v.42 no.11
    • /
    • pp.725-730
    • /
    • 2009
  • Cyclin E1 (CCNE1), a positive regulator of the cell cycle, controls the transition of cells from G1 to S phase. In numerous human tumors, however, CCNE1 expression is frequently dysregulated, while the mechanism leading to its dysregulation remains incompletely defined. Herein, we showed that CCNE1 expression was subject to post-transcriptional regulation by a microRNA miR-16-1. This was evident at protein level of CCNE1 as well as its mRNA level. Further evident by dual luciferase reporter assay revealed that two evolutionary conserved binding sites on 3' UTR of CCNE1 were the direct functional target sites. Moreover, we showed that miR-16-1 induced G0/G1 cell cycle arrest by targeting CCNE1 and siRNA against CCNE1 partially phenocopied miR-16-1-induced cell cycle phenotype whereas substantially rescued anti-miR-16-1- induced phenotype. Together, all these results demonstrate that miR-16-1 plays a vital role in modulating cellular process in human cancers and indicate the therapeutic potential of miR-16-1 in cancer therapy.

In Vitro Anti-Neuroblastoma Activity of Thymoquinone Against Neuro-2a Cells via Cell-cycle Arrest

  • Paramasivam, Arumugam;Raghunandhakumar, Subramanian;Priyadharsini, Jayaseelan Vijayashree;Jayaraman, Gopalswamy
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.16 no.18
    • /
    • pp.8313-8319
    • /
    • 2016
  • We have recently shown that thymoquinone (TQ) has a potent cytotoxic effect and induces apoptosis via caspase-3 activation with down-regulation of XIAP in mouse neuroblastoma (Neuro-2a) cells. Interestingly, our results showed that TQ was significantly more cytotoxic towards Neuro-2a cells when compared with primary normal neuronal cells. In this study, the effects of TQ on cell-cycle regulation and the mechanisms that contribute to this effect were investigated using Neuro-2a cells. Cell-cycle analysis performed by flow cytometry revealed cell-cycle arrest at G2/M phase and a significant increase in the accumulation of TQ-treated cells at sub-G1 phase, indicating induction of apoptosis by the compound. Moreover, TQ increased the expression of p53, p21 mRNA and protein levels, whereas it decreased the protein expression of PCNA, cyclin B1 and Cdc2 in a dose-dependent manner. Our finding suggests that TQ could suppress cell growth and cell survival via arresting the cell-cycle in the G2/M phase and inducing apoptosis of neuroblastoma cells.