INTRODUCTION

AML is the most common malignant myeloid disorder in adults. Relapses are initiated by chemoresistant leukemic cells. DNA damage and repair mechanisms influence not only the genetic predisposition to leukemia but are also very important for refractoriness to treatment.

The aim of this study was to investigate the possible alterations in the gene expression profile in DNA damage signaling pathways in two leukemic cell lines following their exposure to chemotherapeutic agents and verify the findings in AML patients.

METHODS

Kasumi-1 and MV4-11 AML cells were treated with either idarubicin (0.1μΜ) for 6h or cytarabine (1μΜ) for 48h. Dead cells were eliminated from drug-treated cells using the appropriate commercial kit. Gene expression profiling through PCR arrays analysis (RT2Profiler, Qiagen) was performed after RNA extraction from untreated, drug-treated and chemoresistant (live) cells following their exposure to cytotoxic agents. Human DNA Damage Signaling pathway related genes' expression was evaluated and analyzed through RT2Profiler PCR Array data analysis tool. Following our initial results, two genes were selected for further analysis: PPP1R15A and HUS-1 genes' relative expression was evaluated by qRT-PCR analysis with QuantiTect Primer Assays kit (Qiagen) using the 2^-∆∆Ct method. The analysis included 28 de novo AML patients before the onset of the 7+3 combination chemotherapy and 16 healthy donors. Eighteen cases had normal karyotype including 7 with flt3 mutation, 1 case had inv(16) and 9 cases intermediate risk karyotype. Statistics were performed through One Way Anova analysis.

RESULTS

PCR Array analysis after idarubicin and cytarabine treatment of Kasumi-1 cells revealed a significant up-regulation of genes involved in apoptosis, cell cycle, DNA damage and repair, and ATM/ATR signaling. Significant differences in their gene expression patterns were observed between cytarabine-treated Kasumi-1 cells and chemoresistant ones. HUS-1 gene (DSB) was 3x fold up-regulated in cytarabine-treated cells and 0.7x fold down-regulated in chemoresistant cells compared to untreated cells.

Cytarabine and idarubicin treatment of MV4-11 cells led to an up-regulation of genes involved in cell cycle, DNA damage repair, including DSB repair and NER mechanisms. Most importantly, PPP1R15A gene's expression in both cytarabine and idarubicin chemoresistant MV4-11 cells was significantly 4.2x and 2.7x fold up-regulated compared to drug treated cells.

Following these results the expression level of genes PPP1R15A and HUS1 was examined in the bone marrow cells of AML patients in order to verify their association with chemoresistance.

PPP1R15A gene's relative expression was significantly up-regulated in non-responding to induction chemotherapy AML patients compared to responding (median: 2.705 vs. 0.73, p<0.05) and in non-responding to chemotherapy AML patients compared to controls (median: 2.705 vs. 0.577, p<0.01).

HUS1 gene's relative expression was remarkably down-regulated in AML patients compared to controls (median: 1.585 vs. 7.74, p<0.001). This was also observed comparing responding and refractory to chemotherapy AML patients to controls (median: 1.09 vs. 7.74, p<0.001 and 1.585 vs. 7.74, p<0.05, respectively).

CONCLUSIONS

The up-regulation of PPP1R15A gene in chemoresistant MV4-11 cells after treatment with cytotoxic agents is justified since this gene participates in growth arrest and apoptosis in response to DNA damage, negative growth signals and protein malfolding by binding to protein phosphatase PP1, and attenuating the translational elongation of key transcription factors through dephoshorylation of eukaryotic initiation factor 2a(eIF2a). Most importantly the significant increased baseline expression of PPP1R15A in AML chemoresistant patients indicates its involvement in chemoresistance mechanisms and paves the way for targeted treatment.

HUS1 gene's expression was remarkably depressed in de novo AML samples. This gene is required for the optimal ATM/ATR signaling response to DSBs and replication stress forming part of the RAD9A-RAD1-HUS1 (9-1-1) complex functioning as a damage sensor in checkpoint pathway. Therefore the described above reduced expression in AML samples indicates reduced ATM-ATR response to DSBs associated with genetic instability and offers new options for synthetic lethality treatment strategies

Disclosures

Symeonidis:Tekeda: Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Research Funding; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; MSD: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Research Funding; Roche: Membership on an entity's Board of Directors or advisory committees, Research Funding. Pappa:Roche: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Research Funding; Novartis: Honoraria, Research Funding, Speakers Bureau; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Honoraria, Research Funding; Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Research Funding; Celgene / GenesisPharma: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution