scholarly journals Chromatin structure restricts origin utilization when quiescent cells re-enter the cell cycle

2020 ◽  
Author(s):  
Po-Hsuen Lee ◽  
Mary Ann Osley

Abstract Quiescent cells reside in G0 phase, which is characterized by the absence of cell growth and proliferation. These cells remain viable and re-enter the cell cycle when prompted by appropriate signals. Using a budding yeast model of cellular quiescence, we investigated the program that initiated DNA replication when these G0 cells resumed growth. Quiescent cells contained very low levels of replication initiation factors, and their entry into S phase was delayed until these factors were re-synthesized. A longer S phase in these cells correlated with the activation of fewer origins of replication compared to G1 cells. The chromatin structure around inactive origins in G0 cells showed increased H3 occupancy and decreased nucleosome positioning compared to the same origins in G1 cells, inhibiting the origin binding of the Mcm4 subunit of the MCM licensing factor. Thus, quiescent yeast cells are under-licensed during their re-entry into S phase.

2020 ◽  
Author(s):  
Mark C. Johnson ◽  
Geylani Can ◽  
Miguel Santos ◽  
Diana Alexander ◽  
Philip Zegerman

AbstractAcross eukaryotes, checkpoints maintain the order of cell cycle events in the face of DNA damage or incomplete replication. Although a wide array of DNA lesions activates the checkpoint kinases, whether and how this response differs in different phases of the cell cycle remains poorly understood. The S-phase checkpoint for example results in the slowing of replication, which in the budding yeast Saccharomyces cerevisiae is caused by Rad53 kinase-dependent inhibition of the initiation factors Sld3 and Dbf4. Despite this, we show here that Rad53 phosphorylates both of these substrates throughout the cell cycle at the same sites as in S-phase, suggesting roles for this pathway beyond S-phase. Indeed we show that Rad53-dependent inhibition of Sld3 and Dbf4 limits re-replication in G2/M phase, preventing inappropriate gene amplification events. In addition we show that inhibition of Sld3 and Dbf4 after DNA damage in G1 phase prevents premature replication initiation at all origins at the G1/S transition. This study redefines the scope and specificity of the ‘S-phase checkpoint’ with implications for understanding the roles of this checkpoint in the majority of cancers that lack proper cell cycle controls.


2020 ◽  
Author(s):  
Vincent Gaggioli ◽  
Manuela R. Kieninger ◽  
Anna Klucnika ◽  
Richard Butler ◽  
Philip Zegerman

AbstractDuring metazoan development, the cell cycle is remodelled to coordinate proliferation with differentiation. Developmental cues cause dramatic changes in the number and timing of replication initiation events, but the mechanisms and physiological importance of such changes are poorly understood. Cyclin-dependent kinase (CDK) is important for regulating S-phase length in many metazoa, and here we show in the nematode Caenorhabditis elegans that an essential function of CDK during early embryogenesis is to regulate the interactions between three replication initiation factors SLD-3, SLD-2 and MUS-101 (Dpb11/TopBP1). Mutations that bypass the requirement for CDK to generate interactions between these factors is sufficient for viability in the absence of CyclinE/Cdk2, demonstrating that this is a critical embryonic function of this cyclin/CDK complex. Both SLD-2 and SLD-3 are asymmetrically localised in the early embryo and the levels of these proteins inversely correlate with S-phase length. We also show that SLD-2 asymmetry is determined by direct interaction with the polarity protein PKC-3. This study explains the essential function of CDK for replication initiation in a metazoan and provides the first direct molecular mechanism through which polarization of the embryo is coordinated with DNA replication initiation.Author SummaryHow and when a cell divides changes as the cell assumes different fates. How these changes in cell division are brought about are poorly understood, but are critical to ensure that cells do not over-proliferate leading to cancer. The nematode C. elegans is an excellent system to study the role of cell cycle changes during animal development. Here we show that two factors SLD-2 and SLD-3 are critical to control the decision to begin genome duplication. We show that these factors are differently distributed to different cell lineages in the early embryo, which may be a key event in determining the cell cycle rate in these cells. For the first time we show that, PKC-3, a key component of the machinery that determines the front (anterior) from the back (posterior) of the embryo directly controls SLD-2 distribution, which might explain how the polarisation of the embryo causes changes in the proliferation of different cell lineages. As PKC-3 is frequently mutated in human cancers, how this factor controls cell proliferation may be important to understand tumour progression.


2021 ◽  
Vol 22 (10) ◽  
pp. 5195
Author(s):  
Hui Zhang

In eukaryotic cells, DNA replication licensing is precisely regulated to ensure that the initiation of genomic DNA replication in S phase occurs once and only once for each mitotic cell division. A key regulatory mechanism by which DNA re-replication is suppressed is the S phase-dependent proteolysis of Cdt1, an essential replication protein for licensing DNA replication origins by loading the Mcm2-7 replication helicase for DNA duplication in S phase. Cdt1 degradation is mediated by CRL4Cdt2 ubiquitin E3 ligase, which further requires Cdt1 binding to proliferating cell nuclear antigen (PCNA) through a PIP box domain in Cdt1 during DNA synthesis. Recent studies found that Cdt2, the specific subunit of CRL4Cdt2 ubiquitin E3 ligase that targets Cdt1 for degradation, also contains an evolutionarily conserved PIP box-like domain that mediates the interaction with PCNA. These findings suggest that the initiation and elongation of DNA replication or DNA damage-induced repair synthesis provide a novel mechanism by which Cdt1 and CRL4Cdt2 are both recruited onto the trimeric PCNA clamp encircling the replicating DNA strands to promote the interaction between Cdt1 and CRL4Cdt2. The proximity of PCNA-bound Cdt1 to CRL4Cdt2 facilitates the destruction of Cdt1 in response to DNA damage or after DNA replication initiation to prevent DNA re-replication in the cell cycle. CRL4Cdt2 ubiquitin E3 ligase may also regulate the degradation of other PIP box-containing proteins, such as CDK inhibitor p21 and histone methylase Set8, to regulate DNA replication licensing, cell cycle progression, DNA repair, and genome stability by directly interacting with PCNA during DNA replication and repair synthesis.


1999 ◽  
Vol 340 (1) ◽  
pp. 135-141 ◽  
Author(s):  
Parisa DANAIE ◽  
Michael ALTMANN ◽  
Michael N. HALL ◽  
Hans TRACHSEL ◽  
Stephen B. HELLIWELL

The essential cap-binding protein (eIF4E) of Saccharomycescerevisiae is encoded by the CDC33 (wild-type) gene, originally isolated as a mutant, cdc33-1, which arrests growth in the G1 phase of the cell cycle at 37 °C. We show that other cdc33 mutants also arrest in G1. One of the first events required for G1-to-S-phase progression is the increased expression of cyclin 3. Constructs carrying the 5ʹ-untranslated region of CLN3 fused to lacZ exhibit weak reporter activity, which is significantly decreased in a cdc33-1 mutant, implying that CLN3 mRNA is an inefficiently translated mRNA that is sensitive to perturbations in the translation machinery. A cdc33-1 strain expressing either stable Cln3p (Cln3-1p) or a hybrid UBI4 5ʹ-CLN3 mRNA, whose translation displays decreased dependence on eIF4E, arrested randomly in the cell cycle. In these cells CLN2 mRNA levels remained high, indicating that Cln3p activity is maintained. Induction of a hybrid UBI4 5ʹ-CLN3 message in a cdc33-1 mutant previously arrested in G1 also caused entry into a new cell cycle. We conclude that eIF4E activity in the G1-phase is critical in allowing sufficient Cln3p activity to enable yeast cells to enter a new cell cycle.


2001 ◽  
Vol 12 (1) ◽  
pp. 53-62 ◽  
Author(s):  
Matthew R. Alexander ◽  
Mike Tyers ◽  
Mireille Perret ◽  
B. Maureen Craig ◽  
Karen S. Fang ◽  
...  

Exposure of yeast cells to an increase in external osmolarity induces a temporary growth arrest. Recovery from this stress is mediated by the accumulation of intracellular glycerol and the transcription of several stress response genes. Increased external osmolarity causes a transient accumulation of 1N and 2N cells and a concomitant depletion of S phase cells. Hypertonic stress triggers a cell cycle delay in G2 phase cells that appears distinct from the morphogenesis checkpoint, which operates in early S phase cells. Hypertonic stress causes a decrease in CLB2 mRNA, phosphorylation of Cdc28p, and inhibition of Clb2p-Cdc28p kinase activity, whereas Clb2 protein levels are unaffected. Like the morphogenesis checkpoint, the osmotic stress-induced G2 delay is dependent upon the kinase Swe1p, but is not tightly correlated with inhibition of Clb2p-Cdc28p kinase activity. Thus, deletion ofSWE1 does not prevent the hypertonic stress-induced inhibition of Clb2p-Cdc28p kinase activity. Mutation of the Swe1p phosphorylation site on Cdc28p (Y19) does not fully eliminate the Swe1p-dependent cell cycle delay, suggesting that Swe1p may have functions independent of Cdc28p phosphorylation. Conversely, deletion of the mitogen-activated protein kinase HOG1 does prevent Clb2p-Cdc28p inhibition by hypertonic stress, but does not block Cdc28p phosphorylation or alleviate the cell cycle delay. However, Hog1p does contribute to proper nuclear segregation after hypertonic stress in cells that lack Swe1p. These results suggest a hypertonic stress-induced cell cycle delay in G2 phase that is mediated in a novel way by Swe1p in cooperation with Hog1p.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi94-vi94
Author(s):  
Taylor Dismuke ◽  
Chaemin Lim ◽  
Timothy Gershon

Abstract CDK4/6 inhibition is a promising therapy for medulloblastoma, one of the most common malignant pediatric brain tumors. To improve pharmacokinetics, we developed a polyoxazoline nanoparticle-encapsulated formulation of the FDA-approved CDK4/6 inhibitor palbociclib (POx-palbo). We then administered POx-palbo to transgenic medulloblastoma-prone GFAP-Cre/SmoM2 mice, to determine the efficacy and mechanisms of action and resistance. We found that POx-palbo slowed tumor progression, but consistently failed to be curative. Further analysis showed that while CDK4/6 inhibition acutely blocked G1 cells from re-entering the cell cycle, this effect wore off within hours of drug administration. However, flow cytometric analysis of EdU uptake hours after palbociclib demonstrated aberrant S-phase with reduced rate of DNA synthesis. This POx-palbociclib-induced alteration of S-phase progression seems to remain true at later time points even when we observed that palbociclib G1/S inhibition began to decrease. Based on these data, we propose that the combinational therapy of POx-palbociclib and S-phase targeting agents will further improve treatment. Faulty tumor cell cycle progression in the presence of Pox-palbociclib may give increased window to target the S-phase for irreversible cell-cycle exit.


2009 ◽  
Vol 184 (3) ◽  
pp. 409-422 ◽  
Author(s):  
Casey A. Ydenberg ◽  
Mark D. Rose

When yeast cells sense mating pheromone, they undergo a characteristic response involving changes in transcription, cell cycle arrest in early G1, and polarization along the pheromone gradient. Cells in G2/M respond to pheromone at the transcriptional level but do not polarize or mate until G1. Fus2p, a key regulator of cell fusion, localizes to the tip of the mating projection during pheromone-induced G1 arrest. Although Fus2p was expressed in G2/M cells after pheromone induction, it accumulated in the nucleus until after cell division. As cells arrested in G1, Fus2p was exported from the nucleus and localized to the nascent tip. Phosphorylation of Fus2p by Fus3p was required for Fus2p export; cyclin/Cdc28p-dependent inhibition of Fus3p during late G1 through S phase was sufficient to block exit. However, during G2/M, when Fus3p was activated by pheromone signaling, Cdc28p activity again blocked Fus2p export. Our results indicate a novel mechanism by which pheromone-induced proteins are regulated during the transition from mitosis to conjugation.


2019 ◽  
Vol 9 (2) ◽  
pp. 319-323 ◽  
Author(s):  
Li Ping ◽  
Li Mingzhu ◽  
Lü Yuchun

Objective: To explore on the antitumor effect of gefitinib and rapamycin and possible mechanism in normal glucose and high lactic acid microenvironment. Methods: Hela cells are cultured in six conditions: the normal glucose group (NG, glucose 3 mmol/L); the normal glucose + gefitinib group (NGG, glucose 3 mmol/L, gefitinib 2.67 μmol/L); the normal glucose + rapamycin group (NGR, glucose 3 mmol/L, rapamycin 2.67 μmol/L); the high lactate group (NGHL, glucose 3 mmol/L, lactic acid 2.5 mmol/L); the normal glucose + high lactate + gefitinib group (NGHLG, glucose 3 mmol/L, lactic acid 2.5 mmol/L, gefitinib 2.67μmol/L); the normal glucose + high lactate + rapamycin group (NGHLG, glucose 10 mmol/L, lactic acid 2.5 mmol/L, rapamycin 2.67μmol/L). Growth inhibitory rate of Hela cell is determined by CCK-8; Flow cytometry (FCM) is performed to evaluate the cell cycle; The annexin V-phycoerythrin/Propidium Iodide (annexin V-PE/PI) staining combined with flow cytometry is used to examine the cell cycle and apoptosis of Hela cells. Results: Under normal glucose with gefitinib or rapamycin environment, the apoptosis rate of Hela cells is higher than that of the normal glucose group. But the cell apoptosis rate of the gefitinib or rapamycin group decreases in high lactic acid and normal glucose, which is lower than that of the normal glucose and high lactate. Combined with the results of cell cycle, compared with the normal glucose group, percentage of Hela cells in G1/G0 phase increases significantly, the proportion of S phase cells decreases significantly in high lactic acid environment. In the normal glucose and gefitinib environment, Hela cells in G1/G0 phase and S phase are slightly higher than the proportion of normal glucose group, and G2/M phase cells are mild lower than the proportion of normal glucose group. Under the environment of high lactate and normal glucose, the percentage of G1/G0 and S phase cells in the gefitinib increase. As for rapamycin, normal glucose and high lactic acid environment makes cells stay in G1/G0 phase. The presence of rapamycin in the environment of normal sugar and high lactate makes more cells stay in G1/G0 or G2/M phase. Conclusion: Normal glucose and high lactic acid environment is conducive to Hela cell survival, and can promote the expression of EGFR and mTOR. Gefitinib is an antagonist of EGFR and rapamycin is an inhibitor of mTOR.


1996 ◽  
Vol 74 (1) ◽  
pp. 21-28 ◽  
Author(s):  
Thomas J. Logan ◽  
Kelly L. Jordan ◽  
David J. Hall

The E2F1 transcription factor was constitutively expressed in NIH3T3 fibroblasts to determine its effect on the cell cycle. These E2F1 cell lines were not tightly synchronized in G0 phase of the cell cycle following serum starvation, as are normal fibroblasts. Instead, the cells are spread throughout G0 and G1 phase with a portion of the population initiating DNA synthesis. Upon serum stimulation, the remaining cells in G0/G1 begin to enter S phase immediately but with a reduced rate. Constitutive expression of E2F1 appears to primarily affect the G0 phase, since transit of proliferating E2F1 cell lines through G1 phase is the same as control cells. Consistent with a shortened G0 phase, the E2F1 cell lines have a significantly reduced cellular volume. Additionally, the first S phase after serum stimulation, but not subsequent S phases, is nearly doubled in the E2F1 cell lines compared with control cells. Cell lines expressing a deletion mutant of E2F1 (termed E2F1d87), known to significantly affect cell shape, have cell cycle and volume characteristics similar to the E2F1 expressing cells. However, all S phase durations are considerably lengthened and the cells demonstrate delayed growth after plating.Key words: cell cycle, E2F1 transcription factor, G0/G1 phase.


2003 ◽  
Vol 77 (6) ◽  
pp. 3451-3459 ◽  
Author(s):  
Robert F. Kalejta ◽  
Thomas Shenk

ABSTRACT As viruses are reliant upon their host cell to serve as proper environments for their replication, many have evolved mechanisms to alter intracellular conditions to suit their own needs. For example, human cytomegalovirus induces quiescent cells to enter the cell cycle and then arrests them in late G1, before they enter the S phase, a cell cycle compartment that is presumably favorable for viral replication. Here we show that the protein product of the human cytomegalovirus UL82 gene, pp71, can accelerate the movement of cells through the G1 phase of the cell cycle. This activity would help infected cells reach the late G1 arrest point sooner and thus may stimulate the infectious cycle. pp71 also induces DNA synthesis in quiescent cells, but a pp71 mutant protein that is unable to induce quiescent cells to enter the cell cycle still retains the ability to accelerate the G1 phase. Thus, the mechanism through which pp71 accelerates G1 cell cycle progression appears to be distinct from the one that it employs to induce quiescent cells to exit G0 and subsequently enter the S phase.


Sign in / Sign up

Export Citation Format

Share Document