scholarly journals Knockdown of ADORA2A antisense RNA 1 inhibits cell proliferation and enhances imatinib sensitivity in chronic myeloid leukemia

Bioengineered ◽  
2022 ◽  
Vol 13 (2) ◽  
pp. 2296-2307
Author(s):  
Yabo Liu ◽  
Huibo Li ◽  
Yanqiu Zhao ◽  
Dandan Li ◽  
Qian Zhang ◽  
...  
2018 ◽  
Vol 47 (3) ◽  
pp. 981-993 ◽  
Author(s):  
Yu Chen ◽  
Tongtong Wang ◽  
Jing Du ◽  
Yanchun Li ◽  
Xin Wang ◽  
...  

Background/Aims: Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm. Tyrosine kinase inhibitors (TKIs) are commonly used to treat CML; however, drug resistance of CML cells to TKIs has limited their clinical application. Shikonin, a traditional Chinese herb, has long been used to treat leukemia in China, but the roles and related molecular mechanisms of shikonin treatment in CML remain unclear. Here, we aimed to evaluate the effects of shikonin on the proliferation, apoptosis, and migration of K562 cells, a CML cell line. Methods: Firstly, K562 cell proliferation and apoptosis were tested by CCK8 assay and flow cytometry with Annexin V-FITC/PI staining. Cell migration was measured by Transwell migration assay. In addition, western blot was performed to determine the proteins (PI3K, Bax, Bcl-2, cleaved caspase-3, PTEN, p-AKT, AKT, CXCR4, SDF-1, CD44) involved in the mechanism of action of shikonin. Finally, neutrophils from peripheral blood of CML patients were obtained, and cell proliferation and apoptosis were tested by CCK8 assay and flow cytometry. Results: Shikonin reduced the proliferation of K562 cells in a time- and dose-dependent manner and promoted the apoptosis of K562 cells. Moreover, shikonin increased the PTEN level and inactivated the PI3K/AKT signaling pathway, subsequently upregulating BAX in K562 cells. In addition, shikonin could block K562 cell migration via the CXCR4/SDF-1 axis. Finally, shikonin significantly inhibited the proliferation and promoted the apoptosis of neutrophils from CML patients. Conclusion: These results demonstrated that shikonin inhibits CML proliferation and migration and induces apoptosis by the PTEN/PI3K/AKT pathway, revealing the effects of shikonin therapy on CML.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1017-1017
Author(s):  
Michaela Scherr ◽  
Letizia Venturini ◽  
Karin Battmer ◽  
Michael Schaller-Schoenitz ◽  
Daniel Schaefer ◽  
...  

Abstract Micro RNAs (miRNA) are small non-coding RNAs that regulate gene expression by specific hybridization to complementary sequences in the 3′ untranslated region of corresponding mRNAs. Concomitant recruitment of specific multi-protein complexes results either in inhibition of mRNA translation or mRNA degradation. miRNAs are processed in a regulated multi-step process from primary transcripts into mature miRNAs by cellular components which are also at least partially involved in the process of RNA interference (RNAi). Aberrant expression of specific miRNAs has recently been described in human lymphoma and leukemia. In particular, BCR-ABL and c-MYC dependent over-expression of the polycistronic and oncogenic miR-17-92 cluster (encoding miR-17, miR-18a, miR-19a, miR-20a, miR-19b, and miR-92) has been described in chronic myeloid leukemia (CML) cell lines, primary CD34+ cells from CML patients (Venturini et al. 2007), and in lung cancer. In BCR-ABL positive K562 cells, miR-17-92 encoded miRNAs repress luciferase activity in miRNA-specific reporter assays. In addition, lentivirus-mediated over-expression of miR-17-92 increases both cell proliferation and sensitivity to imatinib induced cell death. To analyse the function of individual miRNAs of the miR-17-92 polycistron, we generated lentivirus-based strategies to induce stable miRNA-specific loss- and gain-of function phenotypes for miR-18a, miR-19b, and miR-20a, respectively. Over-expression of miRNAs embedded within miR-30-derived sequences from an internal SFFV-LTR promoter allows isolation of K562 cells with increased miRNA expression. In contrast, expression of complementary oligonucleotides (antagomirs) from a H1 promoter located in the lentiviral 3′LTR can induce stable hypomorphic miRNA-phenotypes. In lentivirally transduced K562 cells, individual silencing of miR-18a, miR-19b, and miR-20a by the corresponding antagomirs (ant-miR-18a, ant-miR-19b, ant-miR-20a) specifically relieves miRNA-mediated reporter gene repression. Correspondingly, inhibition of miRNA-function correlates to reduced ‘miRNA’-amplification by miRNA-specific quantitative RT-PCR. Furthermore, protein expression of E2F-1, a known miR-20 target, is enhanced by lentivirally expressed anti-miR-20 in a dose-dependent manner, whereas over-expression of miR-20a reduces E2F-1 levels. Finally, combined over-expression of specific miRNAs and antagomirs reveals specific induction of cell proliferation by miR-18a but strong inhibition by miR-20a in K562 cells, respectively. In contrast, anti-miR-18a, but not anti-miR-19b, anti-miR-20a, or control antagomirs inhibits proliferation of K562 cells. These data demonstrate individual and complementary functions of miR-17-92 encoded miRNAs in CML and identify potential targets for specific therapeutic intervention on the miRNA level.


2021 ◽  
Vol 11 ◽  
Author(s):  
Minran Zhou ◽  
Xiaolin Yin ◽  
Lixin Zheng ◽  
Yue Fu ◽  
Yue Wang ◽  
...  

BackgroundChronic myeloid leukemia (CML) is a malignant clonal proliferative disease. Once it progresses into the phase of blast crisis (CML-BP), the curative effect is poor, and the fatality rate is extremely high. Therefore, it is urgent to explore the molecular mechanisms of blast crisis and identify new therapeutic targets.MethodsThe expression levels of miR-181d, RBP2 and NF-κB p65 were assessed in 42 newly diagnosed CML-CP patients and 15 CML-BP patients. Quantitative real-time PCR, Western blots, and cell proliferation assay were used to characterize the changes induced by overexpression or inhibition of miR-181d, RBP2 or p65. Luciferase reporter assay and ChIP assay was conducted to establish functional association between miR-181d, RBP2 and p65. Inhibition of miR-181d expression and its consequences in tumor growth was demonstrated in vivo models.ResultsWe found that miR-181d was overexpressed in CML-BP, which promoted leukemia cell proliferation. Histone demethylase RBP2 was identified as a direct target of miR-181d which downregulated RBP2 expression. Moreover, RBP2 inhibited transcriptional expression of NF-κB subunit, p65 by binding to its promoter and demethylating the tri/dimethylated H3K4 region in the p65 promoter locus. In turn, p65 directly bound to miR-181d promoter and upregulated its expression. Therefore, RBP2 inhibition resulting from miR-181d overexpression led to p65 upregulation which further forwarded miR-181d expression. This miR-181d/RBP2/p65 feedback regulation caused sustained NF-κB activation, which contributed to the development of CML-BP.ConclusionsTaken together, the miR-181d/RBP2/p65 feedback regulation promoted CML-BP and miR-181d may serve as a potential therapeutic target of CML-BP.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2712-2712
Author(s):  
Maria Rosaria Ricciardi ◽  
Valentina Salvestrini ◽  
Mattia Forcato ◽  
Roberto Licchetta ◽  
Simone Mirabilii ◽  
...  

Abstract Chronic Myeloid Leukemia (CML) is a stem cell disease sustained by a rare population of kinetic quiescent cells, frequently resistant to tyrosine kinase inhibitors (TKIs). The Bcr-Abl oncogene and the resulting fusion protein, in fact, activates multiple cross-talking signal transduction pathways (STP), such as RAS/MEK/ERK, PI3K/Akt, Wnt and STAT5, potentially contributing to TKIs drug resistance. Since increasing evidence reports the cooperation of numerous STP in the control of cell proliferation and survival in CML, the aim of this project was to analyze, at the protein level, the expression and activation profile of various STP involved in the mechanisms of cell proliferation and survival of CML CD34+ cells, as compared to different sources of normal CD34+ cells. CD34+ cells were purified by immunomagnetic separation from peripheral blood (PB) of 7 newly diagnosed chronic phase (CP) CML patients and compared to the normal counterpart obtained from normal bone marrow of three healthy donors (NBM) and/or from umbilical cord blood (CB) of three donors. The phosphorylation status of 46 different proteins belonging to numerous STP and the expression of 32 proteins of the apoptotic machinery were assessed by using a customized direct phase proteome profiler antibody array. The resulting dots were visualised using ECL and quantified by densitometric analysis. CP-CML samples were obtained from patients with WBC counts ranging between 41,900 to 421,400; Sokal score resulted intermediate in six patients and low in one. The comparison between the phospho-proteomic profile of CP-CML CD34+ cells and NBM CD34+ cells showed that the former are characterized by: 1) lower phosphorylation of STAT2 (p=0.023), Chk-2 (p=0.036), a serine/threonine-protein kinase required for checkpoint-mediated cell cycle arrest, and tyrosine kinases of the Src family - Lck, Fyn, Src, particularly Yes (p=0.04) - involved in the regulation of growth and cell survival; 2) higher phosphorylation of p53, both at Ser15 (p=0.047) and at Ser46 (p=0.039), p70S6 kinase (p=0.035), RSK (p=0.046), a mediator of mitogens- and stress-induced activation of several transcription factors, and Pyk-2 (p=0.036), a tyrosine kinase involved in cell adhesion and migration. The analysis of the 32 apoptotic proteins revealed that CD34+ cells from CP-CML, compared to CD34+ cells from NBM, are characterized by: 1) lower expression of the catalase (p=0.044), an enzyme that protects cells from the toxic effects of hydrogen peroxide and promotes growth of normal and neoplastic cells including myeloid leukemia cells; 2) higher expression of some members of the heat shock proteins family - HSP60 and HSP70 (p=0.033). We then compared CD34+ cells obtained from CP-CML with the other normal CD34+ cell source represented by the CB. The proteomic profile indicated a remarkable similarity between the CD34+ from CP-CML and those from CB. Accordingly, the two normal CD34+ cells sources showed some differences: in particular, as for CP-CML CD34+ cells, those from CB had significantly lower phosphorylation of STAT2 (p=0.026), of Chk-2 (p=0.014) and higher phosphorylation of p53 at Ser15 (p=0.05), compared to NBM CD34+ cells. In summary, we reported that CD34+ cells from CP-CML are characterized by a proteomic and phospho-proteomic profile that promotes quiescence status through the inhibition of proliferation. A striking similarity was found between CD34+ cells obtained from CP-CML and those from CB. The two normal sources of CD34 displayed differences in the activation status of selected proteins. The presence of these additional and complex changes in the signaling network of CP-CML must be taken into account for the investigation on novel targeted therapies. Disclosures: Castagnetti: Novartis Farma : Consultancy, Honoraria; Bristol Myers Squibb : Consultancy, Honoraria. Rosti:Bristol Myers Squibb : Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau; Novartis Pharma: Honoraria, Membership on an entity’s Board of Directors or advisory committees, Research Funding, Speakers Bureau; Roche: Speakers Bureau; Pfizer: Speakers Bureau.


2021 ◽  
Vol 41 (1) ◽  
pp. 79-82
Author(s):  
Jin Wang ◽  
Hu‐Lin Ma ◽  
Wen‐Rong Liu ◽  
Yong Peng ◽  
Jian‐Kang Zhou ◽  
...  

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.


Sign in / Sign up

Export Citation Format

Share Document