Cell cycle and cell size regulation in Down Syndrome cells

Author(s):  
M. Rosner ◽  
A. Kowalska ◽  
A. Freilinger ◽  
A-R. Prusa ◽  
E. Marton ◽  
...  
2013 ◽  
pp. 343-346 ◽  
Author(s):  
Ákos Sveiczer ◽  
Anna Rácz-Mónus
Keyword(s):  

HortScience ◽  
2004 ◽  
Vol 39 (4) ◽  
pp. 868C-868
Author(s):  
Anish Malladi* ◽  
Peter Hirst

Fruit size is a commercially valuable trait. Although several factors are known to affect fruit size in apple, insights into the molecular aspects of its regulation are lacking. Our research aims to understand fruit size regulation using a combination of approaches. Analysis of a large fruited mutant of `Gala', `Grand Gala' (GG), showed that it was 40% heavier than `Gala' at harvest. Increase in size of GG fruit was caused by an increase in the cell size apparent at full bloom. Flow cytometry revealed the presence of multiple levels of ploidy (up to 16C) in GG during early fruit development. Increase in ploidy of GG is hypothesized to be due to endoreduplication, a process normally absent in apple. Endoreduplication is a modification of the cell cycle where DNA replication is not followed by cell division, resulting in increased DNA content accompanied by increased cell size. To understand if the cell cycle is altered in GG, four key cell cycle regulators, MdCDKA1, MdCDKB1, MdCYCB2 and MdCYCD3 have been partially cloned from apple using RT-PCR and RACE. As cell number at the end of the cell division phase is correlated with fruit size at harvest, expression analysis of these genes can provide valuable insights into their role in the regulation of cell number and fruit size. Analysis of cell cycle gene expression in GG may provide key insights into the altered molecular regulation that leads to endoreduplication in the mutant. Parallel approaches being employed to study whether environmental and cultural factors regulate fruit size through an influence on the cell cycle will also be discussed.


2016 ◽  
Vol 113 (51) ◽  
pp. E8238-E8246 ◽  
Author(s):  
Lisa Willis ◽  
Yassin Refahi ◽  
Raymond Wightman ◽  
Benoit Landrein ◽  
José Teles ◽  
...  

Cell size and growth kinetics are fundamental cellular properties with important physiological implications. Classical studies on yeast, and recently on bacteria, have identified rules for cell size regulation in single cells, but in the more complex environment of multicellular tissues, data have been lacking. In this study, to characterize cell size and growth regulation in a multicellular context, we developed a 4D imaging pipeline and applied it to track and quantify epidermal cells over 3–4 d inArabidopsis thalianashoot apical meristems. We found that a cell size checkpoint is not the trigger for G2/M or cytokinesis, refuting the unexamined assumption that meristematic cells trigger cell cycle phases upon reaching a critical size. Our data also rule out models in which cells undergo G2/M at a fixed time after birth, or by adding a critical size increment between G2/M transitions. Rather, cell size regulation was intermediate between the critical size and critical increment paradigms, meaning that cell size fluctuations decay by ∼75% in one generation compared with 100% (critical size) and 50% (critical increment). Notably, this behavior was independent of local cell–cell contact topologies and of position within the tissue. Cells grew exponentially throughout the first >80% of the cell cycle, but following an asymmetrical division, the small daughter grew at a faster exponential rate than the large daughter, an observation that potentially challenges present models of growth regulation. These growth and division behaviors place strong constraints on quantitative mechanistic descriptions of the cell cycle and growth control.


2007 ◽  
Vol 51 (4) ◽  
pp. 642-655 ◽  
Author(s):  
Nathalie Gonzalez ◽  
Frédéric Gévaudant ◽  
Michel Hernould ◽  
Christian Chevalier ◽  
Armand Mouras

2021 ◽  
Author(s):  
Shixuan Liu ◽  
Ceryl Tan ◽  
Chloe Melo-Gavin ◽  
Kevin G. Mark ◽  
Miriam Bracha Ginzberg ◽  
...  

Proliferating animal cells maintain a stable size distribution over generations despite fluctuations in cell growth and division size. This tight control of cell size involves both cell size checkpoints (e.g., delaying cell cycle progression for small cells) and size-dependent compensation in rates of mass accumulation (e.g., slowdown of cellular growth in large cells). We previously identified that the mammalian cell size checkpoint is mediated by a selective activation of the p38 MAPK pathway in small cells. However, mechanisms underlying the size-dependent compensation of cellular growth remain unknown. In this study, we quantified global rates of protein synthesis and degradation in naturally large and small cells, as well as in conditions that trigger a size-dependent compensation in cellular growth. Rates of protein synthesis increase proportionally with cell size in both perturbed and unperturbed conditions, as well as across cell cycle stages. Additionally, large cells exhibit elevated rates of global protein degradation and increased levels of activated proteasomes. Conditions that trigger a large-size-induced slowdown of cellular growth also promote proteasome-mediated global protein degradation, which initiates only after growth rate compensation occurs. Interestingly, the elevated rates of global protein degradation in large cells were disproportionately higher than the increase in size, suggesting activation of protein degradation pathways. Large cells at the G1/S transition show hyperactivated levels of protein degradation, even higher than similarly sized or larger cells in S or G2, coinciding with the timing of the most stringent size control in animal cells. Together, these findings suggest that large cells maintain cell size homeostasis by activating global protein degradation to induce a compensatory slowdown of growth.


2018 ◽  
Author(s):  
Evgeny Zatulovskiy ◽  
Daniel F. Berenson ◽  
Benjamin R. Topacio ◽  
Jan M. Skotheim

Cell size is fundamental to function in different cell types across the human body because it sets the scale of organelle structures, biosynthesis, and surface transport1,2. Tiny erythrocytes squeeze through capillaries to transport oxygen, while the million-fold larger oocyte divides without growth to form the ~100 cell pre-implantation embryo. Despite the vast size range across cell types, cells of a given type are typically uniform in size likely because cells are able to accurately couple cell growth to division3–6. While some genes whose disruption in mammalian cells affects cell size have been identified, the molecular mechanisms through which cell growth drives cell division have remained elusive7–12. Here, we show that cell growth acts to dilute the cell cycle inhibitor Rb to drive cell cycle progression from G1 to S phase in human cells. In contrast, other G1/S regulators remained at nearly constant concentration. Rb is a stable protein that is synthesized during S and G2 phases in an amount that is independent of cell size. Equal partitioning to daughter cells of chromatin bound Rb then ensures that all cells at birth inherit a similar amount of Rb protein. RB overexpression increased cell size in tissue culture and a mouse cancer model, while RB deletion decreased cell size and removed the inverse correlation between cell size at birth and the duration of G1 phase. Thus, Rb-dilution by cell growth in G1 provides a long-sought cell autonomous molecular mechanism for cell size homeostasis.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2721-2721
Author(s):  
Paul Lee ◽  
Rahul Bhansali ◽  
Malini Rammohan ◽  
Nobuko Hijiya ◽  
Shai Izraeli ◽  
...  

Abstract Children with Down syndrome have a spectrum of associated disorders including a 20-fold increased incidence of B-cell acute lymphoblastic leukemia (DS-ALL). Although a number of genetic alterations have been found in this ALL subtype, such as activating mutations in JAK2 and overexpression of CRLF2, the mechanisms by which trisomy 21 promotes the leukemia are largely unknown. Previous studies have implicated chromosome 21 genes HMGN1 and DYRK1A in both malignant and normal lymphopoiesis. DYRK1A is a member of the dual-specificity tyrosine phosphorylation-regulated kinase family that has been well studied in non-hematopoietic tissues. Its targets include proteins that regulate multiple pathways including cell signaling, cell cycle, and brain development. We have previously shown that DYRK1A is a megakaryoblastic leukemia-promoting gene through its negative regulation of NFAT transcription factors. Furthermore, in studies with a conditional Dyrk1a knock-out mouse, we found that the kinase is required for lymphoid, but not myeloid cell development. In developing lymphocytes, Dyrk1a regulates the cell cycle by destabilizing cyclin D3. Consequently, loss of Dyrk1a resulted in the failure of these cells to switch from a proliferative to quiescent phase for subsequent maturation (Thompson et al. J. Exp. Med. 2015 212:953-70). Despite this deficiency in exiting the cell cycle, Dyrk1a-deficient lymphocytes also exhibit impaired proliferation before undergoing apoptosis. These data reveal a critical role for DYRK1A in lymphopoiesis and suggest that it may be a target for therapeutic intervention. We assayed the activity of the highly selective and potent DYRK1 inhibitor, EHT 1610, in multiple ALL cell lines. EHT 1610 inhibited the growth of Jurkat and MHH-CALL-4 cells with EC50s of 0.83mM and 0.49mM, respectively. Next, we treated primary human ALL blasts with EHT 1610 and the less selective DYRK1A inhibitor harmine. Growth of 16 out of 30 specimens, which included DS-ALL, pre-B ALL, and T-ALL, was sensitive to DYRK1A inhibition at doses between 0.5 and 10mM. Of note, growth of 9 of the 11 of the DS-ALL samples was inhibited by EHT 1610. This result indicates that the increased dosage of DYRK1A in DS samples sensitizes the cells to DYRK1A inhibition. To further study the contributions of DYRK1A to normal and malignant lymphopoiesis, we performed phosphoproteomic analysis on primary murine pre-B cells treated with EHT 1610. After 2 hours of EHT 1610 treatment, the cells were collected and analyzed for changes in the phosphoproteome. Phosphorylation of 36 proteins was significantly altered. Bioinformatics analysis led to the identification of a number of notable pathways that appear to be regulated by DYRK1A including cell cycle, cell division and mitosis, RNA metabolism, and JAK-STAT signaling. Differentially phosphorylated proteins included geminin, which is important in cell division and whose loss enhances megakaryopoiesis, and POLR2M, which is intriguing because DYRK1A phosphorylates the CTD of RNA Pol II and binds chromatin at specific sites in glioblastoma cells. Another interesting target is STAT3, which is phosphorylated by DYRK1A on Ser727, a residue whose phosphorylation is required for maximal STAT3 activation. Treatment of murine pre-B cells with EHT 1610 significantly reduced the level of phosphorylation of Ser727 and Tyr705, suggesting that DYRK1A may provide a priming event for STAT3 activation similar to its priming effect on GSK3b phosphorylation. Consistent with a role for JAK/STAT signaling and STAT3 activity, B-ALL cells were highly sensitive to ruxolitinib therapy. Taken together, our study suggests that DYRK1A is a therapeutic target in DS-ALL and likely functions in part by enhancing JAK/STAT signaling. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Author(s):  
Shixuan Liu ◽  
Miriam B. Ginzberg ◽  
Nish Patel ◽  
Marc Hild ◽  
Bosco Leung ◽  
...  

AbstractAnimal cells within a tissue typically display a striking regularity in their size. To date, the molecular mechanisms that control this uniformity are still unknown. We have previously shown that size uniformity in animal cells is promoted, in part, by size-dependent regulation of G1 length. To identify the molecular mechanisms underlying this process, we performed a large-scale small molecule screen and found that the p38 MAPK pathway is involved in coordinating cell size and cell cycle progression. Small cells display higher p38 activity and spend more time in G1 than larger cells. Inhibition of p38 MAPK leads to loss of the compensatory G1 length extension in small cells, resulting in faster proliferation, smaller cell size and increased size heterogeneity. We propose a model wherein the p38 pathway responds to changes in cell size and regulates G1 exit accordingly, to increase cell size uniformity.One-sentence summaryThe p38 MAP kinase pathway coordinates cell growth and cell cycle progression by lengthening G1 in small cells, allowing them more time to grow before their next division.


Sign in / Sign up

Export Citation Format

Share Document