3207 Down-regulated RNF5 (RING finger domain E3 ligase) ubiquitinproteasome pathway related gene expression by resveratrol leads to inhibition of cell proliferation and motility in chronic myeloid leukemia cells

2015 ◽  
Vol 51 ◽  
pp. S651
Author(s):  
C. Biray Avci ◽  
Z. Mutlu ◽  
B. Goker ◽  
C. Caliskan ◽  
O. Ozalp ◽  
...  
Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5369-5369
Author(s):  
Seiichi Okabe ◽  
Yuko Tanaka ◽  
Mitsuru Moriyama ◽  
Akihiko Gotoh

Introduction: ABL tyrosine kinase inhibitors (TKIs) improved outcomes for patients with chronic myeloid leukemia (CML) and Philadelphia chromosome (Ph)-positive leukemia, however, some patients are still resistance to ABL TKIs. One of the most common mechanisms involves point mutations in the kinase domain of BCR-ABL1, however, mechanisms of intrinsic resistance without point mutation of ABL kinase domain are not fully understood. Moreover, ABL TKIs cannot cure the Ph-positive leukemia patients because of leukemia stem cells in the bone marrow niche. Therefore, new approach against leukemia stem cells may improve the outcome of Ph-positive leukemia patients. Hypoxia is an important component of the bone marrow microenvironment. Because oxygen tension plays a key role in driving normal hematopoiesis, leukemia stem cells may be maintained in hypoxic areas of the bone marrow. Materials and methods: In this study, we established ABL TKI-resistant in vitro cell line models (K562 imatinib-R, K562 nilotinib-R, K562 dasatinib-R, K562 ponatinib-R and Ba/F3 T315I). We investigated gene expression profiles in cultured ABL TKI resistant cells and parental cell line, K562 in normoxia and hypoxia condition by DNA microarray. Results: We first investigated gene expression profiles in cultured K562 cells in hypoxia condition. We found gene expression of insulin-like growth factor 1 (IGF1) was increased K562 cells in hypoxia condition by DNA microarray. We next examined ABL TKI resistant cell lines (K562 imatinib-R, K562 nilotinib-R, K562 dasatinib-R, K562 ponatinib-R) in this study. We could not detect the BCR-ABL point mutation in ABL TKI resistant cells. We found gene expression of insulin-like growth factor 1 (IGF1) receptor (IGF1R) was increased ABL TKI resistant K562 cells. IGF1R gene amplification was confirmed by RT-PCR analysis. IGF is tightly regulated by six related IGF-binding proteins (IGFBPs). One of IGFBP, IGFBP5 is related to imatinib sensitivity and resistant in chronic myeloid leukemia (CML) patients (GSE12211). In hypoxia condition, several IGFBPs were also increased in ABL TKI resistant cells. IGF cause intracellular signaling that ultimately results in cellular growth and proliferation. Thus, we initially examined whether addition of IGF1R inhibition could enhance ABL TKIs sensitivity. One of IGF1R inhibitor, linsitinib was inhibited ABL TKI resistant cells and parental cell line, K562 in hypoxia condition. ABL TKI resistant cell lines were more sensitive against linsitinib. Combined treatment of ABL TKI resistant cells and K562 cells with ABL TKIs and linsitinib caused more cytotoxicity than each drug alone in hypoxia condition. Caspase 3/7 activity and cellular cytotoxicity was also increased after ABL TKIs and linsitnib treatment. In the colony formation method, the number of cell colonies were also reduced in hypoxia condition. Intracellular ATP levels have been implicated in vitro as a determinant of cell death by apoptosis. The concentrations of intracellular ATP were reduced after ABL TKIs and linsitinib. We next blocked IGF1R function by small interfering RNA (siRNA). SiRNA transfected cells were reduced cellular proliferation. We also found drug sensitivity of the cells to the imatinib was increased compared to mock-transfected cells. Apoptotic cells and caspase 3/7 activity were increased after imatinib treatment in siRNA transfected cells. Conclusion: The IGF1 pathway is involved in Ph-positive leukemia cells in hypoxia condition and ABL TKI resistant in CML cells. We also provide the promising clinical relevance as a candidate drug for treatment of residual leukemia cells in bone marrow niche which is in hypoxia condition. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Marc García-Montolio ◽  
Cecilia Ballaré ◽  
Enrique Blanco ◽  
Arantxa Gutiérrez ◽  
Sergi Aranda ◽  
...  

Polycomb group (PcG) of proteins are a group of highly conserved epigenetic regulators involved in many biological functions, such as embryonic development, cell proliferation, and adult stem cell determination. PHD finger protein 19 (PHF19) is an associated factor of Polycomb repressor complex 2 (PRC2), often upregulated in human cancers. In particular, myeloid leukemia cell lines show increased levels of PHF19, yet little is known about its function. Here, we have characterized the role of PHF19 in myeloid leukemia cells. We demonstrated that PHF19 depletion decreases cell proliferation and promotes chronic myeloid leukemia (CML) differentiation. Mechanistically, we have shown how PHF19 regulates the proliferation of CML through a direct regulation of the cell cycle inhibitor p21. Furthermore, we observed that MTF2, a PHF19 homolog, partially compensates for PHF19 depletion in a subset of target genes, instructing specific erythroid differentiation. Taken together, our results show that PHF19 is a key transcriptional regulator for cell fate determination and could be a potential therapeutic target for myeloid leukemia treatment.


2015 ◽  
Vol 15 ◽  
pp. S213
Author(s):  
Cansu Caliskan ◽  
Zeynep Mutlu ◽  
Bakiye Goker ◽  
Cagla Kayabasi ◽  
Burcin Tezcanli Kaymaz ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1724-1724 ◽  
Author(s):  
Lin Tong ◽  
Na Xu ◽  
Xuan Zhou ◽  
Jixian Huang ◽  
Wu wan-Er ◽  
...  

Abstract Background: Pyruvate kinase M2 (PKM2) plays an important role in malignant tumor metabolism, and provides tumor cells with enough energy and a lot of metabolic intermediate by mediating Warburg effect. Recently, studies indicate that PKM2 protein can mediate tumor chemoresistance through reprogramming cellular metabolism pathway and participate in chemotherapy resistance mechanisms of many tumor cells. Currently, the problem of imatinib resistance is still a difficult point in the treatment of chronic myeloid leukemia. This study is aimed at investigating the function of PKM2 protein in the metabolism of chronic myeloid leukemia cells and the relationship between PKM2 and drug resistance of chronic myeloid leukemia cells. Methods: We selected the CML primary cells (the cells from new diagnosed patients and refractory patients respectively) and cell lines (KBM5 cells were served as drug-sensitive cells and KBM5 cell with T315I mutation (KBM5-STI) were served as drug-resistant cell) as research objects. We used a low dose PKM2 inhibitor and/or IM treat with CML cells, and detected cell proliferation by CCK-8, apoptosis by flow cytometry. The cell glycolysis level is detected by a real-time cell energy detector, and the results represent the level of glycolysis by the acidification rate. Western blot and RT-qPCR analyzed the protein and mRNA expression level of PKM2 in TKI-sensitive and -resistant primary cells and cell lines. After transfecting control or PKM2 siRNA into KBM5 and KBM5-STI cells for 72hrs, we extracted total RNA and proteins, and determined the mRNA and protein level of PKM2, and the protein level of mTOR、P13K/AKT. Results: PKM2 inhibitor can inhibit KBM5 and KBM5-STI cells growth with IC50 5uM, 6.8uM (P=0.001), respectively, and induce cell apoptosis. Low dose PKM2 inhibitor combined with IM greatly induced KBM5 and KBM5-STI cell growth inhibition and cell death compare to using single drug. PKM2 was higher expressed in the TKI-resistant CML primary cells and cell line KBM5-STI. In the meanwhile, we found that mTOR, HIF-1a were increased in TKI-resistant CML primary cells and cell line too. PKM2 knockdown decreased the cell proliferation and growth, and increased the apoptosis of TKI-resistant cell line after treated with IM. In the mentime, PKM2 knockdown showed significantly lower glycolytic capacity than normal cell line with lower oxygen intake and lacitic acid production, and downregulating with mTOR and P13K/AKT protein expression. Conclusion: Pyruvate kinase M2 (PKM2) acts as an important rate-limiting enzyme in the aerobic glycolytic pathway, and mediates abnormal metabolic pathways which promote tumor cell proliferation, invasion and drug resistance. Compared to the TKI-sensitive primary cell and cell line, PKM2 was increased in the TKI-resistant primary cell and cell line and related to glycolytic level. PKM2 inhibitor can inhibit CML cells growth, induce cell apoptosis, and combined with IM at a low dose can exhibited a synergistic anti-leukemia effect on TKI-resistant cells. Low dose PKM2 inhibitor combined with IM can enhance targeted killing to the CML cells, suggesting that PKM2 protein plays an important role in the formation of drug resistance. After interfering PKM2 protein, it showed a significant downregulated level of glycolysis in the cell line, and the decreased level of P13K/AKT/mTOR signaling pathway. This study demonstrates that PKM2 may be involved in the regulation of energy metabolism in leukemia cells and induce drug resistance. Disclosures No relevant conflicts of interest to declare.


2016 ◽  
Vol 74 (2) ◽  
pp. 277-283 ◽  
Author(s):  
Jingjing Wu ◽  
Bin Wei ◽  
Qian Wang ◽  
Yihan Ding ◽  
Zhikui Deng ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document