restriction point
Recently Published Documents


TOTAL DOCUMENTS

164
(FIVE YEARS 18)

H-INDEX

42
(FIVE YEARS 4)

2021 ◽  
Author(s):  
Deniz Irvali ◽  
Fabian P. Schlottmann ◽  
Prathibha Muralidhara ◽  
Iliya Nadelson ◽  
N. Ezgi Wood ◽  
...  

Eukaryotic cells decide in late G1 whether to commit to another round of genome duplication and division. This point of irreversible cell cycle commitment is a molecular switch termed 'Restriction Point' in mammals and 'Start' in budding yeast. At Start, yeast cells integrate multiple signals such as pheromones, osmolarity, and nutrients. If sufficient nutrients are lacking, cells will not pass Start. However, how the cells respond to nutrient depletion after they have made the Start decision, remains poorly understood. Here, we analyze by live cell imaging how post-Start yeast cells respond to nutrient depletion. We monitor fluorescently labelled Whi5, the cell cycle inhibitor whose export from the nucleus determines Start. Surprisingly, we find that cells that have passed Start can re-import Whi5 back into the nucleus. This occurs when cells are faced with starvation up to 20 minutes after Start. In these cells, the positive feedback loop is interrupted, Whi5 re-binds DNA, and CDK activation occurs a second time once nutrients are replenished. Cells which re-import Whi5 also become sensitive to mating pheromone again, and thus behave like pre-Start cells. In summary, we show that upon starvation the commitment decision at Start can be reversed. We therefore propose that in yeast, as has been suggested for mammalian cells, cell cycle commitment is a multi-step process, where irreversibility in face of nutrient signaling is only reached approximately 20 minutes after CDK activation at Start.


2021 ◽  
Author(s):  
Alan D Stern ◽  
Gregory R Smith ◽  
Luis C Santos ◽  
Deepraj Sarmah ◽  
Xiang Zhang ◽  
...  

Predictive determinants of stochastic single-cell fates have been elusive, even for the well-studied mammalian cell cycle. What drives proliferation decisions of single cells at any given time? We monitored single-cell dynamics of the ERK and Akt pathways, critical cell cycle progression hubs and anti-cancer drug targets, and paired them to division events in the same single cells using the non-transformed MCF10A epithelial line. Following growth factor treatment, in cells that divide both ERK and Akt activities are significantly higher within the S-G2 time window (~8.5-40 hours). Such differences were much smaller in the pre-S-phase, restriction point window which is traditionally associated with ERK and Akt activity dependence, suggesting unappreciated roles for ERK and Akt in S through G2. Machine learning algorithms show that simple metrics of central tendency in this time window are most predictive for subsequent cell division; median ERK and Akt activities classify individual division events with an AUC=0.76. Surprisingly, ERK dynamics alone predict division in individual cells with an AUC=0.74, suggesting Akt activity dynamics contribute little to the decision driving cell division in this context. We also find that ERK and Akt activities are less correlated with each other in cells that divide. Network reconstruction experiments demonstrated that this correlation behavior was likely not due to crosstalk, as ERK and Akt do not interact in this context, in contrast to other transformed cell types. Overall, our findings support roles for ERK and Akt activity throughout the cell cycle as opposed to just before the restriction point, and suggest ERK activity dynamics are substantially more important than Akt activity dynamics for driving cell division in this non-transformed context. Single cell imaging along with machine learning algorithms provide a better basis to understand cell cycle progression on the single cell level.


Author(s):  
Robert F. Brooks

Exit of cells from quiescence following mitogenic stimulation is highly asynchronous, and there is a great deal of heterogeneity in the response. Even in a single, clonal population, some cells re-enter the cell cycle after a sub-optimal mitogenic signal while other, seemingly identical cells, do not, though they remain capable of responding to a higher level of stimulus. This review will consider the origins of this variability and heterogeneity, both in cells re-entering the cycle from quiescence and in the context of commitment decisions in continuously cycling populations. Particular attention will be paid to the role of two interacting molecular networks, namely the RB-E2F and APC/CCDH1 “switches.” These networks have the property of bistability and it seems likely that they are responsible for dynamic behavior previously described kinetically by Transition Probability models of the cell cycle. The relationship between these switches and the so-called Restriction Point of the cell cycle will also be considered.


2021 ◽  
Vol 8 ◽  
Author(s):  
Charline Fagnen ◽  
Ludovic Bannwarth ◽  
Dania Zuniga ◽  
Iman Oubella ◽  
Rita De Zorzi ◽  
...  

In this study, we investigated the dynamics and functional characteristics of the KirBac3.1 S129R, a mutated bacterial potassium channel for which the inner pore-lining helix (TM2) was engineered so that the bundle crossing is trapped in an open conformation. The structure of this channel has been previously determined at high atomic resolution. We explored the dynamical characteristics of this open state channel using an in silico method MDeNM that combines molecular dynamics simulations and normal modes. We captured the global and local motions at the mutation level and compared these data with HDX-MS experiments. MDeNM provided also an estimation of the probability of the different opening states that are in agreement with our electrophysiological experiments. In the S129R mutant, the Arg129 mutation releases the two constriction points in the channel that existed in the wild type but interestingly creates another restriction point.


2021 ◽  
Author(s):  
Han-Wei Shih ◽  
Germain C.M. Alas ◽  
Daria S. Rydell ◽  
Bailin Zhang ◽  
Greyson A. Hamilton ◽  
...  

Transcriptional regulation of differentiation is critical for parasitic pathogens to adapt to environmental changes and regulate transmission. How early signaling transcription factors (TF) activate signal transduction to initiate encystation remains an open question in Giardia. Here, we generate a CasRX-mediated knockdown system, together with an established CRISPRi system to screen early signaling TFs in Giardia lamblia. We identified an early response TF, GARP4 that regulates cyst wall protein (CWP) levels during encystation. Depletion of GARP4 increases encystation efficiency resulting in increased cyst production. Interestingly, cyst viability and CWP1 trafficking are not altered in GARP4 knockdowns, suggesting GARP4 regulates the restriction point controlling the portion of cells that terminally differentiate into cysts. Consistent with previous studies, we find that stimulation of encystation shifts the distribution of cells to the G2/M phase and these cells exhibits higher levels of CWP1, indication that entry into the encystation pathway is cell cycle regulated. Key to this increase of CWP1 in G2/M cells is activation of MYB2, a TF commonly observed during the early phase of encystation in Giardia. Remarkably, activated GARP4 only exhibits in G1/S cells, suggesting it has a role in preventing encystation until G2/M. Furthermore, we demonstrate that depletion of GARP4 activates MYB2 and overexpression of GARP4 represses MYB2. Our findings provide the first molecular mechanism underlying the restriction point regulating differentiation during early signaling of encystation in Giardia lamblia.


Author(s):  
Derouicha Matmour ◽  
Nawel Achachi ◽  
Yassine Mérad ◽  
Houari Toumi

In mammalian cells, proliferation is controlled by the cell cycle, where cyclin-dependent kinases regulate critical checkpoints. CDK4 is considered a highly validated anticancer drug target due to its essential role in regulating cell cycle progression at the G1 restriction point. Our objective is to design novel CDK4 inhibitors using Structure-Based Drug Design and Quantitative Structure-Activity Relationship techniques. We used bioinformatics tools and biological databases. QSAR study of CDK4 inhibitors has given us an idea of the physicochemical features of studied compounds and their correlation with the IC50 activity. The docking study has helped to highlight the molecule key elements to refine in order to get a more potent compound of CDK4.The Molecule under the code 21366124 which has the low IC50= 3 nmole shows the most binding affinity with a score value of ΔG=-9,8 kcal/mol. As prospects, it would be very interesting to synthesize this drug candidate and to test its inhibitory activity on cell culture of breast cancer.


Open Biology ◽  
2020 ◽  
Vol 10 (10) ◽  
pp. 200200
Author(s):  
Eleanor Wendy Trotter ◽  
Iain Michael Hagan

Each approach used to synchronize cell cycle progression of human cell lines presents a unique set of challenges. Induction synchrony with agents that transiently block progression through key cell cycle stages are popular, but change stoichiometries of cell cycle regulators, invoke compensatory changes in growth rate and, for DNA replication inhibitors, damage DNA. The production, replacement or manipulation of a target molecule must be exceptionally rapid if the interpretation of phenotypes in the cycle under study is to remain independent of impacts upon progression through the preceding cycle. We show how these challenges are avoided by exploiting the ability of the Cdk4/6 inhibitors, palbociclib, ribociclib and abemaciclib to arrest cell cycle progression at the natural control point for cell cycle commitment: the restriction point. After previous work found no change in the coupling of growth and division during recovery from CDK4/6 inhibition, we find high degrees of synchrony in cell cycle progression. Although we validate CDK4/6 induction synchronization with hTERT-RPE-1, A549, THP1 and H1299, it is effective in other lines and avoids the DNA damage that accompanies synchronization by thymidine block/release. Competence to return to cycle after 72 h arrest enables out of cycle target induction/manipulation, without impacting upon preceding cycles.


2020 ◽  
Author(s):  
Béla Novák ◽  
John J Tyson

AbstractAs cells pass through each replication-division cycle, they must be able to postpone further progression if they detect any threats to genome integrity, such as DNA damage or misaligned chromosomes. Once a ‘decision’ is made to proceed, the cell unequivocally enters into a qualitatively different biochemical state, which makes the transitions from one cell cycle phase to the next switch-like and irreversible. Each transition is governed by a unique signalling network; nonetheless, they share a common characteristic of bistable behaviour, a hallmark of molecular memory devices. Comparing the cell cycle signalling mechanisms acting at the Restriction Point, G1/S, G2/M and meta-to-anaphase transitions, we deduce a generic network motif of coupled positive and negative feedback loops underlying each transition.


2020 ◽  
Vol 9 (8) ◽  
pp. 2582
Author(s):  
Zihao Chen ◽  
Chunhe Li

The cell cycle, essential for biological functions, experiences delicate spatiotemporal regulation. The transition between G1 and S phase, which is called the proliferation–quiescence decision, is critical to the cell cycle. However, the stability and underlying stochastic dynamical mechanisms of the proliferation–quiescence decision have not been fully understood. To quantify the process of the proliferation–quiescence decision, we constructed its underlying landscape based on the relevant gene regulatory network. We identified three attractors on the landscape corresponding to the G0, G1, and S phases, individually, which are supported by single-cell data. By calculating the transition path, which quantifies the potential barrier, we built expression profiles in temporal order for key regulators in different transitions. We propose that the two saddle points on the landscape characterize restriction point (RP) and G1/S checkpoint, respectively, which provides quantitative and physical explanations for the mechanisms of Rb governing the RP while p21 controlling the G1/S checkpoint. We found that Emi1 inhibits the transition from G0 to G1, while Emi1 in a suitable range facilitates the transition from G1 to S. These results are partially consistent with previous studies, which also suggested new roles of Emi1 in the cell cycle. By global sensitivity analysis, we identified some critical regulatory factors influencing the proliferation–quiescence decision. Our work provides a global view of the stochasticity and dynamics in the proliferation–quiescence decision of the cell cycle.


2020 ◽  
Author(s):  
Robert F. Brooks

AbstractThe Restriction Point (R) in the mammalian cell cycle is regarded as a critical transition in G1 when cells become committed to enter S phase even in the absence of further growth factor stimulation. Classic time-lapse studies by Zetterberg and Larsson suggested that the acquisition of growth factor independence (i.e. passage of R) occurred very abruptly 3-4 hours after mitosis, with most cell cycle variability arising between R and entry into S phase. However, the cycle times of the post-R cells that continued on to mitosis after serum step-down without perturbation were far less variable than the control cells with which they were compared. A re-analysis of the data, presented here, shows that when the timing of R and entry in mitosis are compared for the same experiments, the curves are superimposable and statistically indistinguishable. This indicates that the data are compatible with the timing of R contributing to much of the overall variability in the cell cycle, contrary to the conclusions of Zetterberg and colleagues.


Sign in / Sign up

Export Citation Format

Share Document