Cell Cycle Synchronization and Flow Cytometry Analysis of Mammalian Cells

Author(s):  
Naoko Yoshizawa-Sugata ◽  
Hisao Masai
2021 ◽  
Author(s):  
◽  
Reem Hanna

<p>Peloruside A, a natural product isolated from the marine sponge Mycale hentscheli, is a microtubule-stabilising agent that has a similar mechanism of action to the anticancer drug paclitaxel and is cytotoxic to cultured mammalian cells. Peloruside appears to bind to a distinct site on mammalian tubulin that is different from that of the taxoid-site drugs. Because of the high sequence homology between yeast and mammalian tubulin, Saccharomyces cerevisiae (S. cerevisiae) was used as a model organism to characterise the peloruside-binding site with the aim of advancing our understanding about this site on mammalian tubulin. Wild type S. cerevisiae (BY4741) was sensitive to peloruside at uM concentrations; however, a strain that lacks the mad2 (Mitotic Arrest Deficient 2) gene showed increased sensitivity to the drug at much lower uM concentrations. This gene is a component of the spindle-assembly checkpoint complex that delays the onset of anaphase in cells with defects in mitotic spindle assembly. The main aims of this project were to define the binding site of peloruside A using yeast tubulin to see if microtubule function and/or morphology is altered in yeast by peloruside, and to identify any secondary drug targets "friends of the target" through chemical genetic interactions profiling (Homozygous deletion profiling microarray). Site-directed mutagenesis was used to mutate two conserved amino acids (A296T; R306H) known to confer resistance to peloruside in mammalian cells. Based on a published computer model of the peloruside binding site on mammalian tubulin, we also mutated three other amino acids, two that were predicted to affect peloruside binding (Q291M and N337L), and one that was predicted to affect laulimalide binding but have little affect on peloruside binding (V333W). We also included a negative control that was predicted to have no effect on peloruside binding (R282Q) and would affect epothilone binding. We found that of the six point mutations, only Q291M failed to confer resistance in yeast and instead it increased the inhibition to the drug. Using a bud index assay, confocal microscopy, and flow cytometry, 40-50 uM peloruside was shown to block cells in G2/M of the cell cycle, confirming a direct action of the drug on microtubule function. Homozygous profiling (HOP) microarray analysis of a deletion mutant set of yeast genes was also carried out to identify gene products that interact with peloruside in order to link the drug to specific networks or biochemical pathways in the cells. From site-directed mutagenesis, we concluded that peloruside binds to yeast B-tubulin in the region predicted by the published model of the binding site, and therefore mapping the site on yeast tubulin could provide useful information about the mammalian binding site for peloruside. The bud index, flow cytometry, and confocal microscopy experiments provided further evidence that peloruside interacts with yeast tubulin. From HOP we found that peloruside has roles in the cell cycle, as expected, and has effects on protein transport, secretion, cell wall synthesis, and steroid biosynthesis pathways.</p>


Cells ◽  
2018 ◽  
Vol 7 (11) ◽  
pp. 199 ◽  
Author(s):  
Xiaolan Chen ◽  
Hongjia Ouyang ◽  
Zhijun Wang ◽  
Biao Chen ◽  
Qinghua Nie

It is well known that fibroblast growth factor receptor 2 (FGFR2) interacts with its ligand of fibroblast growth factor (FGF) therefore exerting biological functions on cell proliferation and differentiation. In this study, we first reported that the FGFR2 gene could generate a circular RNA of circFGFR2, which regulates skeletal muscle development by sponging miRNA. In our previous study of circular RNA sequencing, we found that circFGFR2, generated by exon 3–6 of FGFR2 gene, differentially expressed during chicken embryo skeletal muscle development. The purpose of this study was to reveal the real mechanism of how circFGFR2 affects skeletal muscle development in chicken. In this study, cell proliferation was analyzed by both flow cytometry analysis of the cell cycle and 5-ethynyl-2′-deoxyuridine (EdU) assays. Cell differentiation was determined by analysis of the expression of the differentiation marker gene and Myosin heavy chain (MyHC) immunofluorescence. The results of flow cytometry analysis of the cell cycle and EdU assays showed that, overexpression of circFGFR2 accelerated the proliferation of myoblast and QM-7 cells, whereas knockdown of circFGFR2 with siRNA reduced the proliferation of both cells. Meanwhile, overexpression of circFGFR2 accelerated the expression of myogenic differentiation 1 (MYOD), myogenin (MYOG) and the formation of myotubes, and knockdown of circFGFR2 showed contrary effects in myoblasts. Results of luciferase reporter assay and biotin-coupled miRNA pull down assay further showed that circFGFR2 could directly target two binding sites of miR-133a-5p and one binding site of miR-29b-1-5p, and further inhibited the expression and activity of these two miRNAs. In addition, we demonstrated that both miR-133a-5p and miR-29b-1-5p inhibited myoblast proliferation and differentiation, while circFGFR2 could eliminate the inhibition effects of the two miRNAs as indicated by rescue experiments. Altogether, our data revealed that a novel circular RNA of circFGFR2 could promote skeletal muscle proliferation and differentiation by sponging miR-133a-5p and miR-29b-1-5p.


2006 ◽  
Vol 18 (2) ◽  
pp. 209
Author(s):  
M. A. Ramírez ◽  
E. Pericuesta ◽  
M. Pérez-Crespo ◽  
R. Fernández-González ◽  
P. N. Moreira ◽  
...  

Normally the majority of mammalian cells, including murine embryonic stem (mES) cells, are immersed in a low oxygen environment (hypoxia); however, mES are generally cultured in an atmosphere containing 21% O2 (normoxia). Such conditions may not be the most appropriate for mES propagation. We have tested the effects of hypoxia and culture on either feeder fibroblasts or gelatin substrate on mES cell growth and spontaneous differentiation. Two ES cell lines (R1 129/Sv from the laboratory of A. Nagy and MAR B6D2 F1 generated in our laboratory) were cultured under two different oxygen tensions (5 and 21%), and on two different substrates, 0.1% gelatin or murine embryonic fibroblasts (mEF). Cell cycle, cell proliferation, mRNA expression of pluripotency and differentiation markers, as well as spontaneous differentiation to cardiomyocytes, were monitored. For cell proliferation measurements, mES after 7 days of culture at the different conditions were labeled with 5-(and-6)-carboxyfluorescein diacetate succinimidyl ester, and cultured for up to three more days. Cells were then harvested for flow cytometry analysis every 24 h after labeling (Cell TraceTM CFSE Cell Proliferation Kit; Molecular Probes, Inc., Eugene, OR, USA). For cell cycle analysis, cells grown on mEF under the two different oxygen tensions were fixed after 10 days of culture, and then stained with propidium iodide/Triton-X-100 for flow cytometry analysis (Current Protocols in Cytometry, Chap. 7, 2001). The spontaneous differentiation of embryoid bodies [formed by ES cells in the absence of leukemia inhibitory factor (LIF)] to cardiomyocytes was also monitored. For mRNA expression of pluripotency (Nanog, Oct-3/4, Rex1, GENESIS, FGFR-4, TERF1, Cx43, and GLUT1) and differentiation markers (GATA4, GATA2, AFP, Msx-1, Brachyury, and Myf5), RT-PCR analysis was performed on mES cells from Day 0 to Day 10. Under hypoxia conditions, the proliferation of both types of mES cells was greater than under normoxia, independent of substrate used, and a higher number of apoptotic cells was detected. Moreover, only under normoxia conditions did mES cells lose the expression of pluripotency markers GENESIS and GLUT1. In addition, under lower oxygen tension, the rate of differentiation to beating cardiomyocytes was significantly lower on the feeder layer than that under normoxia (11.9% vs. 28.1%; P = 0.012). The feeder layer supported significantly higher cardiomyocyte formation when compared to 0.1% gelatin at 21% O2 (28.1% vs. 8.3%; P < 0.001). Our results show that normoxia may not be the most appropriate condition for mES cell propagation and that hypoxic culture may be necessary to maintain full pluripotency of mES cells.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 329-329
Author(s):  
Sathisha Upparahallivenkateshaiah ◽  
Khan Sharmin ◽  
Ling Wen ◽  
Rakesh Bam ◽  
Xin Li ◽  
...  

Abstract Abstract 329 Accumulating evidence indicate that cellular metabolism and bi-products also play important roles in signaling associated with tumor cell proliferation, cell cycle, survival and drug resistance. The overall goal of the study was to molecularly characterize MM cells grown in the supportive bone marrow (BM) of clinically relevant SCID-hu or SCID-rab models. MM cells from 22 patients were engrafted in experimental animals. Following establishment of the disease as determined by increased production of circulating human immunoglobulins over a period of 2–4 months, MM cells were isolated from the implanted bones and subjected to global gene expression profile (GEP). Based on stringent criteria (e.g. p<0.05, >2 folds) we identified commonly overexpressed or underexpressed genes in post-engrafted MM cells compared to pre-engrafted cells from the same patients. Among the top upregulated genes we identified several factors associated with lipid metabolism including FABP5 (fatty acid-binding protein 5), SCD (stearoyl CoA desaturase 1), FADS1 (fatty acid desaturase 1) and SLC27A5 (a fatty acid transporter). Clinical GEP data of newly diagnosed patients from Total Therapy program at our institute revealed upregulation of these genes in high risk patients. We further sought to unravel the role of SCD in MM since it has been previously implicated in tumorigenesis and specific inhibitors are being developed for clinical use. SCD (encodes SCD1), is a rate-limiting enzyme responsible for synthesis of monounsaturated fatty acids. We hypothesized that while nutrient unsaturated fatty acids sufficiently satisfy requirement of most normal cells, growing MM cells demand higher content of these lipids for formation of new membrane phospholipids and immediate energy; therefore, inhibiting SCD1 may suppress MM cell survival and proliferation. Small-molecule inhibitor of SCD1 (BioVision) suppressed growth of 5 MM lines dose dependently; 72 hours IC50 ranged between 1μM (p<0.0006) and 2.5 μM (p<0.0001). At 1 μM the SCD1 inhibitor reduced MM cell proliferation by 70±4% (p<0.002) using thymidine incorporation assay and increased number of apoptotic MM cells from 10±1% in control cells to 27±8% in SCD1 inhibitor-treated cells (p<0.03), using annexin V/PI flow cytometry analysis. This inhibitor also disrupted cell cycle progression in MM cell lines as determined by flow cytometry analysis of DNA content. The Akt/mTOR and AMPK pathways, albeit opposing functions, are known central integrators of cellular metabolism and proliferation signaling. SCD1 inhibitor reduced phosphorylated AKT and increased phosphorylated AMPK in MM cells assessed by Western Blot. For in vivo experiments in SCID-rab mice, SCD1 inhibitor was constantly administered (1.25 μg/hour) by osmotic pumps directly connected to the implanted bones that had been engrafted with luciferase-expressing H929 MM cells (6 mice/group). SCD1 inhibitor suppressed MM growth by 60% (p<0.01) assessed by live-animal imaging and measurement of circulating levels of human immunoglobulins in mice sera. These findings suggest that intracellular modulators of lipid metabolism such as SCD1 are induced in MM cells by the supportive BM and mediate signals linking cellular metabolism, survival and proliferation. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 50 (5) ◽  
pp. 453-463 ◽  
Author(s):  
Israel Torres-Castro ◽  
César N. Cortés-Rubio ◽  
Guadalupe Sandoval ◽  
Edmundo Lamoyi ◽  
Carlos Larralde ◽  
...  

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Shan Yang ◽  
Kai Zeng ◽  
Ling Luo ◽  
Wang Qian ◽  
Zhiqiang Wang ◽  
...  

1992 ◽  
Vol 21 (1) ◽  
pp. 39-42 ◽  
Author(s):  
G. Lizard ◽  
P. Roignot ◽  
L. Dusserre-Guion ◽  
F. Morlevat ◽  
D. Michiels-Marzais ◽  
...  

eLife ◽  
2019 ◽  
Vol 8 ◽  
Author(s):  
Eszter Zavodszky ◽  
Ramanujan S Hegde

We have used misfolded prion protein (PrP*) as a model to investigate how mammalian cells recognize and degrade misfolded GPI-anchored proteins. While most misfolded membrane proteins are degraded by proteasomes, misfolded GPI-anchored proteins are primarily degraded in lysosomes. Quantitative flow cytometry analysis showed that at least 85% of PrP* molecules transiently access the plasma membrane en route to lysosomes. Unexpectedly, time-resolved quantitative proteomics revealed a remarkably invariant PrP* interactome during its trafficking from the endoplasmic reticulum (ER) to lysosomes. Hence, PrP* arrives at the plasma membrane in complex with ER-derived chaperones and cargo receptors. These interaction partners were critical for rapid endocytosis because a GPI-anchored protein induced to misfold at the cell surface was not recognized effectively for degradation. Thus, resident ER factors have post-ER itineraries that not only shield misfolded GPI-anchored proteins during their trafficking, but also provide a quality control cue at the cell surface for endocytic routing to lysosomes.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jia-Lu Zheng ◽  
Shuang-Shuang Wang ◽  
Ke-Ping Shen ◽  
Lei Chen ◽  
Xiao Peng ◽  
...  

Abstract Background Ursolic acid (UA) is an anti-cancer herbal compound. In the present study, we observed the effects of UA on anchorage-dependent and -independent growth of human colorectal cancer (CRC) RKO cells. Methods RKO cells were cultured in conventional and detached condition and treated with UA. Cell viability was evaluated by CCK-8 assay. Cell cycle was analyzed by flow cytometry. Apoptosis was identified by Hoechst 33258 staining and flow cytometry analysis. Activities of caspases were measured by commercial kits. Reactive oxygen species (ROS) was recognized by DCFH-DA fluorescent staining. Anoikis was identified by EthD-1 fluorescent staining and flow cytometry analysis. Expression and phosphorylation of proteins were analyzed by western blot. Results UA inhibited RKO cell viability in both a dose- and time-dependent manner. UA arrested the cell cycle at the G0/G1 phase, and induced caspase-dependent apoptosis. UA inhibited Bcl-2 expression and increased Bax expression. In addition, UA up-regulated the level of ROS that contributed to UA activated caspase-3, − 8 and − 9, and induced apoptosis. Furthermore, UA inhibited cell growth in a detached condition and induced anoikis in RKO cells that was accompanied by dampened phosphorylation of FAK, PI3K and AKT. UA also inhibited epithelial-mesenchymal transition (EMT) as indicated by the down-regulation of N-Cad expression and up-regulation of E-Cad expression. Conclusions UA induced caspase-dependent apoptosis, and FAK/PI3K/AKT singling and EMT related anoikis in RKO cells. UA was an effective anti-cancer compound against both anchorage-dependent and -independent growth of RKO cells.


Sign in / Sign up

Export Citation Format

Share Document