Ultrasonic Assisted Cerium Oxide/Graphene Oxide Hybrid: Preparation, Anti-proliferative, Apoptotic Induction and G2/M Cell Cycle Arrest in HeLa Cell Lines

2019 ◽  
Vol 30 (7) ◽  
pp. 2666-2676 ◽  
Author(s):  
J. Saranya ◽  
B. S. Sreeja ◽  
G. Padmalaya ◽  
S. Radha ◽  
T. Manikandan
Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4360-4360
Author(s):  
SIN Chun-fung ◽  
Timothy Ming-hun Wan ◽  
Aarmann Anil Mohinani Mohan ◽  
Yinxia Qiu ◽  
Anan Jiao

Abstract T lymphoblastic leukaemia (T-ALL) is an aggressive haematological malignancy with poor outcome, especially for relapse/refractory disease. Early T- cell precursor acute lymphoblastic leukaemia (ETP-ALL) is a recently identified subtype of T-ALL with worse treatment outcome compared with other subtypes of T-ALL and treatment options are limited. T-ALL frequently harbors genetic aberrations leading to cell cycle dysregulation and it is one of the major molecular pathogenesis of T-ALL. WEE1 is a protein kinase that is responsible for inhibiting mitosis with unrepaired damaged DNA via inactivating CDK1. WEE1 is highly express in adult T-ALL and its overexpression is associated with adverse prognosis in various cancers. Inhibiting WEE1 expression is a novel approach of therapy. Bortezomib is a 26S proteosome inhibitor and it is FDA approved for treating plasma cell myeloma and mantle cell lymphoma. Bortezomib had been demonstrated therapeutic efficacy in clinical setting for relapse/refractory paediatric T-ALL and B-ALL when combined with chemotherapy. Despite its therapeutic efficacy in clinical studies, the mechanism of action of Bortezomib in T-ALL remain uncertain. The role of Bortezomib in cell cycle modulation had not been established in T-ALL. Moreover, it had not been demonstrated that the effect of Bortezomib in WEE1 expression in T-ALL. Here, we present our study that demonstrated the therapeutic efficacy of Bortezomib in treating T-ALL via cell cycle modulation and downregulation of WEE1 by Bortezomib. T-ALL cell lines including MOLT16, MOLT4, LOUCY and CEM were used in the study. Cell viability was measured by trypan blue. Apoptosis and cell cycle analysis were measured by flow cytometry. Western blot of WEE1, p53, cyclin B1, p21 and p27 were performed. Our result showed that Bortezomib reduce the cell viability of T-ALL cell lines in dose and time-dependent manner. Bortezomib was also sensitive towards LOUCY, a T-ALL cell line with ETP-ALL phenotype. It implied that Bortezomib could be a promising therapy for ETP-ALL. Bortezomib also triggered apoptosis in various T-ALL and the effect of apoptosis was more pronounced after 72 hours of treatment when compared with 24-hour. Again, Bortezomib was able to induce apoptosis in LOUCY cell line. G2/M cell cycle arrest was observed in various T-ALL upon treatment of Bortezomib. The effect on cell cycle modulation was also observed in LOUCY cell line. The protein expression of p21 and p27 were increased after the treatment of Bortezomib. The level of cyclin B1 was increased also. There was upregulation of p53 after Bortezomib treatment. Strikingly, the protein expression level of WEE1 was reduced. The findings of WEE1 downregulation by Bortezomib is a novel findings. We also showed that Bortezomib downregulate WEE1 mRNA expression by quantitative PCR. Our study showed that Bortezomib is active against T-ALL cell lines, including ETP-ALL cell line, LOUCY and modulates cell cycle with G2/M arrest. Bortezomib had been shown to increase the level of p21, p27 and cyclin B1 and induced G2/M cell cycle arrest in glioblastoma cells. However, studies on cell cycle modulation by Bortezomib in T-ALL are scarce. Here, we demonstrated Bortezomib stabilized p21, p27 and upregulation of cyclin B1 in T-ALL as well, which could account for the G2/M cell cycle arrest. We first showed that downregulation of WEE1 after treatment with Bortezomib, in protein level as well as in mRNA level. Recent study showed that inhibition of WEE1 is a novel target of therapy in T-ALL. WEE1 is upregulated in T-ALL to prevent entry of mitosis with unrepaired damaged DNA. The downregulation of WEE1 by Bortezomib as showed by our study could reverse its effect and leads to apoptosis of leukaemic cells. In summary, our study provides the insight on mechanism of action of Bortezomib in modulating cell cycle in T-ALL. Moreover, it is the first study to demonstrate WEE1 downregulation by Bortezomib in T-ALL. These findings not only enhance our understanding of mechanism of action of Bortezomib in T-ALL, but also rationalized the use of certain synergistics combination therapy with Bortezomib in treating T-ALL, e.g., chemotherapeutic agents, PARP inhibitors which could damage DNA of leukaemic cells. Further research is needed to explore those combination therapy in T-ALL and molecular mechanism of downregulation of WEE1 by Bortezomib in T-ALL. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
saranya J ◽  
BS Sre ◽  
M Arivanandan ◽  
K Bhuvaneswari ◽  
S Sherin ◽  
...  

Abstract Using the ultrasonic approach, we produced a morphology involving cerium oxide/ Zinc oxide/graphene oxide (CeO2/ZnO/GO) nanocomposite-based system. The developed nanocomposite was examined using X-Ray diffraction (XRD), Fourier transform infrared spectroscopy (FTIR), and field emission scanning electron microscopy (FESEM). The average crystallite size was found to be 11.44 nm, as determined by XRD. FTIR analysis was used to confirm the existence of functional groups. FESEM was used to verify the morphological properties of CeO2/ZnO/GO. The micromorphology of CeO2/ZnO/GO nanocomposite reveals a smoother sheet-like structure. In addition, using an antiproliferative assay test, the developed nanosystem was evaluated for its scavenging anti-cancer capability against HeLa cell lines at various doses and incubation intervals. In our investigation, the effective IC50 concentration was reported to be 62.5 µg/ml at 72 h. Further, the developed nanosystem was evaluated for its killing efficacy against normal cell line. To identify apoptosis-associated alterations of cell membranes throughout the apoptosis process, a dual acridine orange/ethidium bromide (AO/EB) fluorescent staining was done using CeO2/ZnO/GO nanocomposite at three specific concentrations. The quantitative analysis was carried out using flow cytometry (FACS study) to determine the cell cycle during which the greatest number of HeLa cells were destroyed. According to the results of the FACS investigation, maximum cell cycle has taken place in P2, P4.As a result, the newly designed CeO2/ZnO/GO hybrid has demonstrated improved anti-cancer efficacy against the HeLa cell line, making it a better therapeutic agent for cervical cancer detection.


2006 ◽  
Vol 66 (6) ◽  
pp. 3238-3247 ◽  
Author(s):  
Maria Grazia Villani ◽  
Valentina Appierto ◽  
Elena Cavadini ◽  
Arianna Bettiga ◽  
Alessandro Prinetti ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5170-5170
Author(s):  
Ebenezer David ◽  
Jonathan L Kaufman ◽  
Jack Arbiser ◽  
Sagar Lonial

Abstract Background: Multiple myeloma is an incurable malignancy of plasma cells. While conventional and novel therapies have improved the outcome of patients with myeloma, new therapies and therapeutic targets are needed. NADPH oxidase represents a potential therapeutic target, as NADPH oxidase inhibitors block transduction of signals from cell surface receptors and the PI3 kinase pathway that are dependent on reactive oxygen species (ROS). We tested the effect of the NADPH oxidase inhibitor, Imipramine Blue (IB), on myeloma cell growth and activation of apoptosis in cell lines and in primary human myeloma cells. Methods: IB was synthesized by refluxing imipramine and michlers ketone in the presence of phosphorus oxychloride. MTT assays for myeloma cells (MM.1S, MM.1R, RPMI8226, and U266) were used to evaluate the cell viability. AnnexinV staining and cell cycle analysis was done by flow cytometry to assess the level of apoptosis and analyze cell cycle arrest. Western blotting was performed using antibodies to analyse the impact of IB on intracellular signaling targets. Results: Myeloma cells were treated with increasing concentrations of IB for 72 hrs. Cell viability assays demonstrated that treatment of myeloma cell lines with IB resulted in an 80% decrease in cell growth using concentrations of IB between 6 and 10 uM. To determine the cause of growth inhibition and further mechanism of action, assays for apoptosis and cell cycle were performed. Annexiv V staining demonstrated that myeloma cell lines underwent brisk and rapid apoptosis. Procaspases (caspase 8, 9, 3 and PARP) were extensively cleaved from concentrations as low as 0.5uM to 10uM of IB at the 48 hour timepoint. Also, extensive DNA damage was evident based on the elevated levels of phos-p53 and GADD45 in MM.1S cells. The cell cycle profile indicated that IB induces both G2/M cell cycle arrest in MM.1S cells as well as in RPMI8226 cell lines. In addition, IB overcomes the growth advantage by cytokines IL-6 and IGF1 in MM.1S cells. Primary tumor cells obtained from myeloma patients demonstrated a significant cell killing following IB exposure. Given single agent activity, we then combined IB with other active anti-myeloma agents. Combining IB either with the proteasome inhibitor bortezomib, perifosine (an AKT inhibitor) or honokiol, a natural extract of the magnolia flower (Ishitsuka; Blood; 2005) demonstrated significant increase in the level of apoptosis favoring the combination in myeloma cells. Additionally, BAY11-7082 is an inhibitor of cytokine-induced IKB-α phosphorylation enhanced the level of apoptosis in combination with IB in the U266 myeloma cell line which is relatively less sensitive to IB, suggesting inhibition of NF-KB pathway is involved in the IB mediated myeloma cell death. Conclusion: Imipramine Blue has a potential to induce both apoptosis and G2/M cell cycle arrest in myeloma cell lines and in primary myeloma cells. The cellular response seems to be mainly mediated through pathways independent of PI3 kinase pathway, causing extensive DNA damage, cell cycle arrest, and apoptosis.


Sign in / Sign up

Export Citation Format

Share Document