scholarly journals ALK Inhibitors-Induced M Phase Delay Contributes to the Suppression of Cell Proliferation

Cancers ◽  
2020 ◽  
Vol 12 (4) ◽  
pp. 1054
Author(s):  
Sirajam Munira ◽  
Ryuzaburo Yuki ◽  
Youhei Saito ◽  
Yuji Nakayama

Anaplastic lymphoma kinase (ALK), a receptor-type tyrosine kinase, is involved in the pathogenesis of several cancers. ALK has been targeted with small molecule inhibitors for the treatment of different cancers, but absolute success remains elusive. In the present study, the effects of ALK inhibitors on M phase progression were evaluated. Crizotinib, ceritinib, and TAE684 suppressed proliferation of neuroblastoma SH-SY5Y cells in a concentration-dependent manner. At approximate IC50 concentrations, these inhibitors caused misorientation of spindles, misalignment of chromosomes and reduction in autophosphorylation. Similarly, knockdown of ALK caused M phase delay, which was rescued by re-expression of ALK. Time-lapse imaging revealed that anaphase onset was delayed. The monopolar spindle 1 (MPS1) inhibitor, AZ3146, and MAD2 knockdown led to a release from inhibitor-induced M phase delay, suggesting that spindle assembly checkpoint may be activated in ALK-inhibited cells. H2228 human lung carcinoma cells that express EML4-ALK fusion showed M phase delay in the presence of TAE684 at about IC50 concentrations. These results suggest that ALK plays a role in M phase regulation and ALK inhibition may contribute to the suppression of cell proliferation in ALK-expressing cancer cells.

2018 ◽  
Vol 19 (12) ◽  
pp. 4014 ◽  
Author(s):  
Daiki Okumura ◽  
Mari Hagino ◽  
Akane Yamagishi ◽  
Yuichiro Kaibori ◽  
Sirajam Munira ◽  
...  

Cell division is the process by which replicated chromosomes are separated into two daughter cells. Although regulation of M phase has been extensively investigated, not all regulating factors have been identified. Over the course of our research, small molecules were screened to identify those that regulate M phase. In the present study, the vascular endothelial growth factor receptor (VEGFR) inhibitors A83-01, SU4312, and Ki8751 were examined to determine their effects on M phase. Treatment of HeLa S3 cells with these inhibitors suppressed cell proliferation in a concentration-dependent manner, and also suppressed Akt phosphorylation at Ser473, a marker of Akt activation. Interestingly, cleaved caspase-3 was detected in Adriamycin-treated cells but not in inhibitor-treated cells, suggesting that these inhibitors do not suppress cell proliferation by causing apoptosis. A cell cycle synchronization experiment showed that these inhibitors delayed M phase progression, whereas immunofluorescence staining and time-lapse imaging revealed that the M phase delay was accompanied by misalignment of chromosomes and rotation of the mitotic spindle. Treatment with the Mps1 inhibitor AZ3146 prevented the SU4312-induced M phase delay. In conclusion, the VEGFR inhibitors investigated here suppress cell proliferation by spindle assembly checkpoint-induced M phase delay, via misalignment of chromosomes and rotation of the mitotic spindle.


Planta Medica ◽  
2017 ◽  
Vol 83 (14/15) ◽  
pp. 1149-1158 ◽  
Author(s):  
Elisabetta Corradi ◽  
Nadine Schmidt ◽  
Nathalie Räber ◽  
Maria De Mieri ◽  
Matthias Hamburger ◽  
...  

AbstractPhenolic constituents of Salix reticulata (Salicaceae) and antiproliferative activity of an extract and individual compounds were investigated in immortalized human non-tumorigenic keratinocytes (HaCaT). A MeOH extract from aerial parts afforded several flavonoids, including luteolin and apigenin glycosides (2–5 and 9) and catechin (1), two procyanidin fractions, and the phenolic glucosides picein (6), triandrin (7), and salicortin (8). In an adenosine triphosphate assay, the MeOH extract reduced cell viability by approximately 60 % at a concentration of 100 µg/mL. Cell proliferation was assessed with a BrdU incorporation ELISA assay. The extract inhibited proliferation of HaCaT cells in a concentration-dependent manner, with approximately 50 % inhibition at 100 µg/mL. In time-lapse assays, the extract showed distinct inhibitory effects on cell migration at concentrations of 12.5, 25, and 50 µg/mL. The activity of selected constituents was also determined. Luteolin-7-O-β-glucuronide (3) significantly inhibited cell proliferation at concentrations of 10 and 50 µM. In contrast, luteolin-7-O-β-glucopyranoside (2) and a procyanidin fraction (P1) had only weak effects, while picein (6) and salicortin (8) did not affect cell proliferation. Luteolin-7-O-β-glucuronide (10 µM) and, to a lesser extent, the procyanidin fraction (10 µg/mL) also inhibited cell migration.


2020 ◽  
Vol 7 (Supplement_1) ◽  
pp. S659-S660
Author(s):  
Brett Baker

Abstract Background The rise in resistance to existing antimicrobials has prompted a need for the development of novel antibiotics. Microbion has identified a novel compound, pravibismane, with potent broad spectrum anti-infective and anti-biofilm activity. Methods Here we used a variety of assays, including Bacterial Cytological Profiling (BCP), to analyze pravibismane in E.coli to gain insight into its likely mechanism of action (MOA). The BCP profile of pravibismane suggested it rapidly shut down cell growth, potentially by turning off cellular gene or protein expression. This was confirmed using a plasmid based GFP induction assay in E.coli tolC that showed pravibismane strongly reduced expression of GFP. The kinetics, reversibility and MOA of pravibismane was further characterized by using time-lapse microscopy, wash out experiments and measurements of both membrane potential and relative intracellular ATP levels. Results We found that pravibismane acts rapidly (within 30 mins) to completely halt cell growth rather than causing immediate cell lysis such as that observed with non-specific cell damaging agents bleach or detergent. Inhibitor wash out experiments in which cells were exposed to pravibismane for 2 hours, washed to remove the compound, and then observed using time-lapse microscopy revealed that the effect of pravibismane is reversible and that cells recovered 8-12 hrs after removing the compound. Wash out experiments with an E.coli tolC strain carrying a plasmid with an IPTG inducible GFP demonstrated that transcription and translation ultimately resumed in most cells after washout. The bioenergetics of the membrane was measured using DiBAC 4(5), a membrane potential sensitive dye which can enter depolarized cells, which revealed that pravibismane caused depolarization of the membrane within 30 mins of exposure in a concentration dependent manner. Finally, a luciferase assay determined pravibismane reduced ATP levels (resulting in decreased luminescence) within 15 mins of exposure in a concentration dependent manner unlike antibiotic controls that had modest or no effect on luminescence. Conclusion Our results suggest that pravibismane acts rapidly to disrupt cellular bioenergetics, resulting in the immediate cessation of cell growth and protein expression. Disclosures Brett Baker, M.Sc., D.C., Microbion Corporation (Board Member, Employee)


2022 ◽  
Vol 12 (4) ◽  
pp. 873-877
Author(s):  
Dongqian Xie ◽  
Zhicheng Gao ◽  
Mei Liu ◽  
Defeng Wang

Metformin is shown to have hypoglycemic effects. However, the relationship between metformin’s intervention in FFA-induced endoplasmic reticulum stress-mediated insulin resistance (IR) and insulin β-cell apoptosis under high-glucose condition remains unclear. Our study intends to assess their relationship. Human pancreatic β-cells were treated with metformin and cell proliferation and IR were detected by MTT assay along with detection of Wnt/β-catenin signaling by RT-PCR, cell cycle and apoptosis by flow cytometry. Metformin inhibited β cell proliferation which was mediated by FFA-induced endoplasmic reticulum stress in a time-dependent and dose-dependent manner as well as induced cell cycle arrest at G2/M phase. In addition, metformin inhibited β-catenin signaling activation and decreased the expression of c-myc, Dvl-2, survivin, Dvl-3, GSK-3β (p-ser9) and promoted GSK-3 (p-tyr216) and Axin-2 expression. In conclusion, metformin inhibits Wnt/β-catenin signaling and promotes FFA to induce endoplasmic reticulum stress, thereby mediating pancreatic β-cells behaviors.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 5052-5052 ◽  
Author(s):  
Haiwen Huang ◽  
De Pei Wu ◽  
Anskar Y.H. Leung ◽  
Raymond Liang ◽  
Albert K.W. Lie

Abstract Peroxisome proliferation activated receptor-g (PPAR-g) belongs to the family of nuclear hormone receptors (NHRs), it is normally expressed in adipocytes, adrenal gland, spleen and liver. Recently it was reported that PPAR-g can also be found in tumor tissues. Activation of PPAR-g by its ligands has potential anti-neoplastic effects in a variety of human malignancies, including leukemia through inhibition of cell proliferation, induction of apoptosis and terminal differentiation. The ligands of PPAR-g may represent a promising, novel therapeutic approach for certain human malignancies. The thiazolidinedione (TZD) class drug rosiglitazone (RGZ), one of synthetic ligands of PPAR-g, is currently used for the treatment of type 2 diabetes. In this study, we cultured myeloma cell line U266 with different concentration of rosiglitazone, as well as combined with dexamethasone. Cell proliferation was measured by [3H] thymidine incorporation after 48h incubation. Rosiglitazone was found to generate inhibition of cell proliferation on U266 cells in a dose-dependent manner. Cell cycle analysis by flow cytometry showed that rosiglitazone can arrested U266 cells in G0/G1 phase and the G2/M phase cells were significantly decreased compared to controls. We also studied the effect of rosiglitazone on expression of different anti-apoptosis protein FLIP and survivin by RT-PCR, the result revealed that rosiglitazone can also decrease the FLIP and survivin expression. Furthermore, exposure to rosiglitazone can induce the decreased caspase-3 activity in U266 cells, which was associated with apoptosis induction. When rosiglitazone was combined with dexamethasone, the data demonstrated that cell growth inhibition and apoptosis they exerted was much greater than they were used alone, the decreased expression of FLIP and survivin and decreased caspase-3 activity were also greater than when rosiglitazone was used alone. Based on these findings, we suppose that rosiglitazone alone and in combination with dexamethasone holds promise as novel therapy for myeloma.


2021 ◽  
Author(s):  
Nusrat Sharmeen ◽  
Chris Law ◽  
Cunle Wu

Polarization or directional growth is a major morphological change that occurs in yeast cells during pheromone response to mate with the opposite partner. In the pheromone signaling pathway, the adaptor Ste50 is required to bind MAP3K Ste11 for proper polarization; cells lacking Ste50 are impaired in polarization. Direct involvement of Ste50 in the polarization process has not been explored systematically. Here, we used single-cell fluorescent time-lapse microscopy to characterize Ste50 involvement in the establishment of cell polarity. We found early localization of Ste50 patches on the cell cortex that mark the point of shmoo initiation, these polarity sites move, and patches remain associated with the growing shmoo tip in a pheromone concentration-dependent manner until shmoo maturation. By quantitative analysis we show that polarization corelates with the rising levels of Ste50 enabling rapid individual cell responses to pheromone that corresponds to a critical level of Ste50 at the initial G1 phase. Suggesting Ste50 to be a pheromone responsive gene. We exploited the quantitative differences in the pattern of Ste50 expression to corelate with the cell-cell phenotypic heterogeneity showing Ste50 involvement in the cellular differentiation choices. Taken together, these findings present spatiotemporal localization of Ste50 during yeast polarization, suggesting that Ste50 is a component of the polarisome, and plays a critical role in regulating the polarized growth of shmoo during pheromone response.


2014 ◽  
Vol 998-999 ◽  
pp. 160-163
Author(s):  
Qin Ru ◽  
Xiang Tian ◽  
Yu Xiang Wu ◽  
Kai Yue ◽  
Lin Chen ◽  
...  

Previous studies demonstrated that 20(S)-ginsenoside Rg3 (20S-Rg3) could effectively inhibit tumor cell proliferation as well as K+ channel currents expressed in xenopus oocytes. However, the effect of 20S-Rg3 on the growth of human glioma cells and the ion channels expressed in tumor cells was rarely reported in the literature. In the present study, we investigated the effect and the underlying mechanism of 20S-Rg3 on cell proliferation and apoptosis of human glioma U87-MG cells. In vitro results exhibited that 20S-Rg3 had potent cytotoxic effect and significantly inhibited the proliferation of U87-MG cells in a dose-and time-dependent manner. Typical arrest at G2/M phase was induced, and the apoptosis rate of U87-MG cells was significantly higher in the 20S-Rg3 treatment group than in the control group. Electrophysiological results showed that 80 μmol/L 20S-Rg3 substantially inhibited voltage-gated K+ currents of U87-MG cells. Together, these results suggest that the suppression of voltage-gated K+ currents might play an important role in the 20S-Rg3-induced cell death, and these new findings provide useful data for further study of the antitumor effect of 20S-Rg3.


Blood ◽  
2001 ◽  
Vol 98 (4) ◽  
pp. 995-1002 ◽  
Author(s):  
Maria Koziolkiewicz ◽  
Edyta Gendaszewska ◽  
Maria Maszewska ◽  
C. A. Stein ◽  
Wojciech J. Stec

Many reports indicate different nonantisense yet sequence-specific effects of antisense phosphorothioate oligonucleotides. Products of enzymatic degradation of the oligonucleotides can also influence cell proliferation. The cytotoxic effects of deoxyribonucleoside-5′-phosphates (dNMPs) and their 5′-phosphorothioate analogs, deoxyribonucleoside-5′-monophosphorothioates (dNMPSs) on 4 human cell types (HeLa, HL-60, K-562, and endothelial cells) were examined, and the effects were correlated with the catabolism of these compounds. The results indicate that differences in cytotoxicity of dNMPs or dNMPSs in these cells depend upon different activity of an ecto-5′-nucleotidase. It has also been found that dNMPSs stimulate proliferation of human umbilical vein endothelial cells and HL-60 cells in a concentration-dependent manner. This stimulation might be caused by the binding of deoxynucleoside-5′-phosphorothioates to as-yet unidentified nucleotide receptor(s) at the cell surface.


2002 ◽  
Vol 11 (1) ◽  
pp. 39-45 ◽  
Author(s):  
Fadia R. Homaidan ◽  
Marwan E. El-Sabban ◽  
Iman Chakroun ◽  
Mirvat El-Sibai ◽  
Ghassan S. Dbaibo

Background: In inflammatory bowel disease (IBD), cytokine levels (such as interleukin-1 (IL-1)) are elevated. We have shown previously that IL-1 activates phospholipid signaling pathways in intestinal epithelial cells (IEC), leading to increased ceramide levels.Aim: To determine whether ceramide induces apoptosis in IEC.Methods: Apoptosis was evaluated by annexin-Vbinding or Hoechst nuclear staining. Levels of bcl-2, bcl-x, bax, p53 and p21 were determined by Western blotting, and cell cycle analysis was determined by flow cytometry.Results: IL-1 increased ceramide accumulation in a time-dependent and concentration-dependent manner with a peak response at 4 h, with [IL-1] = 30 ng/ml. Neither IL-1 nor ceramide induced apoptosis in IEC, but they increased bcl-2 levels and decreased bax and p21 levels without affecting bcl-x and p53 levels. They also caused a slight but significant increase in the G2/M phase. These data suggest a role for ceramide in IBD and suggest a possible mechanism for the enhanced tumorigenic activity in IBD patients.


Sign in / Sign up

Export Citation Format

Share Document