scholarly journals Cell-Cycle Gene Expression in Lovastatin-Induced Medulloblastoma Apoptosis

Author(s):  
Wei Wang ◽  
Robert J.B. Macaulay

Background:3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase is a key rate-limiting enzyme in the mevalonate pathway, which generates precursors both for cholesterol biosynthesis and for the production of nonsteroidal mevalonate derivatives that are involved in a number of growth-regulatory processes. We have reported that lovastatin, a competitive inhibitor of HMG-CoA reductase, not only inhibits medulloblastoma proliferation in vitro, but also induces near-complete cell death via apoptosis. The mechanism of this phenomenon is unclear. Possible involvement of changes in expression of certain cell-cycle related genes led us to study some of them in more detail.Methods:Medulloblastoma cell lines were exposed in vitro to lovastatin, and the effects of gene expression changes were studied using RT-PCR, antisense oligonucleotide, DNA electrophoresis and Western blotting analysis.Results:1) Levels of total Ras gene mRNA and individual Ras gene mRNA are stable in lovastatin treatment in all examined medulloblastoma cell lines. 2) Blocking c-myc gene over-expression does not enhance medulloblastoma cell sensitivity to lovastatin. 3) Following lovastatin treatment, p16 expression exhibits no change, but pronounced increases of p27KIP1 protein are observed in all examined cell lines. Lovastatin induces pronounced increases of p21WAF1 protein only in Daoy and UW228, but not in D283 Med and D341 Med. 4) Following lovastatin treatment, increased p53 protein is detected only in D341 Med, and bax protein is unchanged in all cell lines.Conclusion:Lovastatin-induced growth inhibition and apoptosis in medulloblastoma are not dependent on the regulation of Ras and c-myc gene expression, but may be mediated by p27KIP1 gene expression. Lovastatin-induced apoptosis in medulloblastoma is probably p53 independent, but p53 and p21WAF1 gene expression may also mediate anti-proliferative effects of lovastatin on specific medulloblastoma cell lines.

Author(s):  
Wei Wang ◽  
Robert J.B. Macaulay

Background:3-hydroxy-3-methylglutaryl coenzyme A reductase (HMG-CoA reductase) is a key rate-limiting enzyme in the mevalonate pathway, which generates precursors for cholesterol biosynthesis and the production of non-steroidal mevalonate derivatives that are involved in a number of growth-regulatory processes. We have reported that lovastatin, a competitive inhibitor of HMG-CoA reductase, not only inhibits medulloblastoma proliferationin vitro, but also induces near-complete cell death via apoptosis. The present study explores some of the pathways which may be involved in lovastatin-induced apoptosis.Methods:Medulloblastoma cell lines were exposedin vitroto lovastatin with or without mevalonate, and document the effects using morphology, flow cytometry, DNA electrophoresis and Northern analysis.Results:1) Mevalonate prevents apoptosis when co-incubated with lovastatin, or when administered to lovastatin-pretreated cells. 2) Mevalonate restores the lovastatin-arrested cell cycle, allowing S phase entry. 3) Mevalonate does not prevent lovastatin-induced apoptosis after a critical duration of lovastatin pretreatment. For cell lines Daoy and UW228 this was 24 hours, and for D283 Med and D341 Med it was 48 hours. 4) Increases in HMG-CoA reductase mRNA levels induced by lovastatin are abrogated by co-incubation with lovastatin and mevalonate.Conclusion:These results confirm that lovastatin inhibition of this enzyme results in blockage of the mevalonate pathway, and that such a block is a critical step in the mechanism of lovastatin-induced apoptosis.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1005-1005
Author(s):  
Rosa Diaz ◽  
Jonathan M Flanagan ◽  
Thad A Howard ◽  
Russell E. Ware

Abstract Abstract 1005 Hydroxyurea has emerged over the past decade as an effective therapeutic agent for patients with sickle cell anemia (SCA). However, drug dosing and hematological responses can be highly variable; both %HbF response and maximum tolerated dose (MTD) vary widely among patients with SCA who receive hydroxyurea treatment. To obtain further insight into the cellular and molecular pathways, as well as genetic factors that might influence the hydroxyurea MTD, K562 erythroleukemia cells were exposed to hydroxyurea in vitro, to create cell lines that were highly drug tolerant to doses ranging from 250μM to 1500μM. Cell lines had dose-response curves that exhibited clear drug tolerance; naïve K562 showed 50% proliferation in the presence of 250μM hydroxyurea, while tolerant cell lines showed >90% proliferation at the same dose as measured by the BrdU Cell Proliferation Assay. In addition, the tolerant lines showed normal and equivalent progression through cell cycle by flow cytometry cell cycle analysis. After 15 weeks of continuous exposure, cells were harvested and mRNA microarray expression profiles were analyzed for naïve K562 (no hydroxyurea exposure) and cell lines tolerant to 500, 1000, or 1500μM hydroxyurea. Gene expression was measured on Affymetrix U133 Plus 2.0 chips. Differential expression between sample groups was determined using ANOVA, and p-values were corrected for multiple testing using the Benjamin-Hochberg false discovery rate (FDR) method to identify genetic profiles and genes consistently increased or decreased compared to naïve K562 cells. Using a threshold of 2-fold change compared to untreated cells and a false discovery rate <5%, a total of 864 genes were significantly altered in hydroxyurea tolerant cells, including 337 genes whose expression consistently correlated with increasing hydroxyurea dose (Pearson correlation p<.001). The PANTHER classification system was used to group genes into categories based on molecular functions. Of the genes that correlated significantly with increasing hydroxyurea dosing (n=337), there were 181 up-regulated genes and 156 down-regulated genes that had molecular functions including catalytic activity, binding, transcription regulator activity and transporter activity. Genes with transporter activity included SLC6A19, ATP6VOD1, ABCG2, ATP6V1B2 and KCNN4. Other genes of interest based on function included RRM2, PLS3, KCNAB2, UBE2A and SRI. Real-time quantitative reverse transcription (RT)-PCR then quantified the expression of 20 candidate genes to verify the accuracy of the microarray expression data. The next steps will include correlation of these findings with clinical data, specifically early reticulocyte mRNA expression and hydroxyurea MTD values obtained from children with SCA enrolled in the prospective Hydroxyurea Study of Long-term Effects (HUSTLE, NCT00305175). These data document that continuous in vitro exposure of K562 cells to hydroxyurea leads to tolerant cell lines that feature substantial changes in gene expression. Altered expression of certain genes present in erythroid cells including RRM2 and membrane transporters represent compensatory changes in response to hydroxyurea exposure, and may help explain the variability in hydroxyurea MTD observed among patients with SCA. Disclosures: Off Label Use: Hydroxyurea is not FDA approved for pediatric sickle cell patients. Howard:Baylor College of Medicine: Employment.


2010 ◽  
Vol 8 (10) ◽  
pp. 1344-1357 ◽  
Author(s):  
Jens Bunt ◽  
Talitha G. de Haas ◽  
Nancy E. Hasselt ◽  
Danny A. Zwijnenburg ◽  
Jan Koster ◽  
...  

2013 ◽  
Author(s):  
Ricardo Bonfim-Silva ◽  
Thais Valeria A. C. Pimentel ◽  
Elvis T. Valera ◽  
Carlos Alberto Scrideli ◽  
Fernando S. Ramalho ◽  
...  

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 34-35
Author(s):  
Qiang Liu ◽  
Amit Subedi ◽  
Samantha Yao ◽  
Changjiang Xu ◽  
Veronique Voisin ◽  
...  

The main barrier to curing acute myeloid leukemia (AML) is disease relapse, which occurs due to therapy resistance and persistence of leukemic stem cells (LSCs) after conventional induction chemotherapy. Thus drug discovery efforts must focus on identifying agents that effectively target LSCs and not just bulk blasts. To this end, we employed a multi-parametric stemness screen of 1200 bioactive small molecules to identify drugs targeting LSCs, based on reduction of the stem cell compartment of a functionally-characterized hierarchical AML model (OCI-AML-8227) assessed by flow cytometry. In this cell line, self-renewing LSCs are restricted to the CD34+CD38- fraction. The screen identified a number of compound classes with the potential to antagonize LSC properties, including those already in clinical use for AML as well as classes of compounds whose effects in AML have not been previously reported (Figure 1A). Top hits were further validated based on treatment-induced alteration of the expression profile of 104 LSC genes (LSC104) differentially expressed between LSC+ and LSC- fractions of primary AML, which captures stemness properties. The LSC17 score, which is strongly associated with survival and response to standard therapy in AML, was derived from the LSC104 gene set. Notably, all Polo-like kinase 1 (PLK1) inhibitors in the library were identified as top hits in the screen. In vitro treatment of OCI-AML-8227 cells with PLK1 inhibitors over 3 days selectively inhibited the CD34+CD38- fraction enriched in LSCs (Figure 1B), and decreased correlation of gene expression to the LSC104 signature in bulk cells (Figure 1C). Together, these data support a role for PLK1 in regulating leukemic stemness, and we prioritized this class of compounds for validation studies. PLK1 is an important regulator of cell cycle and its best studied role is in the regulation of mitotic entry. However, PLK1 is expressed in and likely plays an important role in all phases of the cell cycle. For instance, PLK1 has been described to regulate cilia disassembly at G0/1. The PLK1 inhibitor volasertib was previously tested in a Phase III trial against AML in combination with low-dose cytarabine (LDC) for elderly patients not eligible for induction chemotherapy. In this trial, although efficacy was observed, significant toxicity in the volasertib+LDC treatment arm resulted in poor survival outcomes for this group of patients. We evaluated the toxicity of volasertib treatment in vitro against two hierarchical AML cell lines (OCI-AML-8227 and OCI-AML-21) as well as normal cord blood (CB). Similar to CB, self-renewing stem cells for these two AML cell lines are restricted to the CD34+CD38- fraction. Treatment with volasertib at 20nM over three days resulted in significantly more cytotoxicity to the AML cell lines compared to CB (Figure 1D), especially in the CD34+CD38- compartment, suggesting that a therapeutic window exists. To evaluate the effects of PDK1 inhibitors against LSCs in vivo, we treated mice bearing AML patient xenografts with single-agent volasertib at a low dose (10mg/kg twice weekly for 4 weeks by oral gavage) starting 4 weeks post-transplant. The gene expression profile for 2 of 4 samples tested showed decreased correlation to the LSC104 signature after volasertib treatment, supporting an effect on stemness (Figure 1E). Volasertib treatment significantly reduced AML engraftment in 4 of 7 samples (Figure 1F). To evaluate the effect of volasertib on LSCs in primary treated mice, we performed secondary transplantation at limiting doses. Volasertib treatment significantly reduced LSC frequency in 2 of 4 samples tested (Figure 1G). Notably, sample AML5 showed a 31.8-fold reduction in LSC frequency compared to controls (p = 0.039) despite no significant reduction in bulk engraftment in primary treated mice, suggesting that volasertib may selectively target LSCs in this sample. In conclusion, our data indicate that the PLK1 inhibitor volasertib, identified as a top hit in a stemness-based drug screen, can target LSCs and decrease stemness properties in some primary AML samples. These findings support further studies of the potential of PLK1 inhibitors for the treatment of AML. Figure Disclosures Wang: Trilium Therapeutics: Patents & Royalties.


2008 ◽  
Vol 8 (3) ◽  
pp. 172-179 ◽  
Author(s):  
M. Bacolod ◽  
S. Lin ◽  
S. Johnson ◽  
N. Bullock ◽  
M. Colvin ◽  
...  

2020 ◽  
Vol 2 (1) ◽  
Author(s):  
Megan Rose Paul ◽  
Yuchen Huo ◽  
Andrea Liu ◽  
Jacqueline Lesperance ◽  
Alexandra Garancher ◽  
...  

Abstract Background Identifying mechanisms of medulloblastoma recurrence is a key to improving patient survival, and targeting treatment-resistant subpopulations within tumors could reduce disease recurrence. Expression of the granulocyte colony-stimulating factor receptor (G-CSF-R, CD114) is a potential marker of cancer stem cells, and therefore we hypothesized that a subpopulation of medulloblastoma cells would also express CD114 and would demonstrate chemoresistance and responsiveness to G-CSF. Methods Prevalence of CD114-positive (CD114+) cells in medulloblastoma cell lines, patient-derived xenograft (PDX) tumors, and primary patient tumor samples were assessed by flow cytometry. Growth rates, chemoresistance, and responses to G-CSF of CD114+ and CD114-negative (CD114−) cells were characterized in vitro using continuous live cell imaging and flow cytometry. Gene expression profiles were compared between CD114+ and CD114− medulloblastoma cells using quantitative RT-PCR. Results CD114+ cells were identifiable in medulloblastoma cell lines, PDX tumors, and primary patient tumors and have slower growth rates than CD114− or mixed populations. G-CSF accelerates the growth of CD114+ cells, and CD114+ cells are more chemoresistant. The CD114+ population is enriched when G-CSF treatment follows chemotherapy. The CD114+ population also has higher expression of the CSF3R, NRP-1, TWIST1, and MYCN genes. Conclusions Our data demonstrate that a subpopulation of CD114+ medulloblastoma cells exists in cell lines and tumors, which may evade traditional chemotherapy and respond to exogenous G-CSF. These properties invite further investigation into the role of G-CSF in medulloblastoma therapy and methods to specifically target these cells.


Sign in / Sign up

Export Citation Format

Share Document